Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Breast Cancer Res Treat ; 166(1): 277-287, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28730340

RESUMEN

PURPOSE: A number of studies have tested the hypothesis that breast cancer patients with low-activity CYP2D6 genotypes achieve inferior benefit from tamoxifen treatment, putatively due to lack of metabolic activation to endoxifen. Studies have provided conflicting data, and meta-analyses suggest a small but significant increase in cancer recurrence, necessitating additional studies to allow for accurate effect assessment. We conducted a retrospective pharmacogenomic analysis of a prospectively collected community-based cohort of patients with estrogen receptor-positive breast cancer to test for associations between low-activity CYP2D6 genotype and disease outcome in 500 patients treated with adjuvant tamoxifen monotherapy and 500 who did not receive any systemic adjuvant therapy. METHODS: Tumor-derived DNA was genotyped for common, functionally consequential CYP2D6 polymorphisms (*2, *3, *4, *6, *10, *41, and copy number variants) and assigned a CYP2D6 activity score (AS) ranging from none (0) to full (2). Patients with poor metabolizer (AS = 0) phenotype were compared to patients with AS > 0 and in secondary analyses AS was analyzed quantitatively. Clinical outcome of interest was recurrence free survival (RFS) and analyses using long-rank test were adjusted for relevant clinical covariates (nodal status, tumor size, etc.). RESULTS: CYP2D6 AS was not associated with RFS in tamoxifen treated patients in univariate analyses (p > 0.2). In adjusted analyses, increasing AS was associated with inferior RFS (Hazard ratio 1.43, 95% confidence interval 1.00-2.04, p = 0.05). In patients that did not receive tamoxifen treatment, increasing CYP2D6 AS, and AS > 0, were associated with superior RFS (each p = 0.0015). CONCLUSIONS: This population-based study does not support the hypothesis that patients with diminished CYP2D6 activity achieve inferior tamoxifen benefit. These contradictory findings suggest that the association between CYP2D6 genotype and tamoxifen treatment efficacy is null or near null, and unlikely to be useful in clinical practice.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Citocromo P-450 CYP2D6/genética , Genotipo , Polimorfismo Genético , Adulto , Anciano , Alelos , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/epidemiología , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Variantes Farmacogenómicas , Pronóstico , Análisis de Supervivencia , Tamoxifeno/uso terapéutico , Resultado del Tratamiento
2.
Pharmacogenomics J ; 17(6): 521-527, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-27549341

RESUMEN

Discovery of clinical and genetic predictors of exemestane pharmacokinetics was attempted in 246 postmenopausal patients with breast cancer enrolled on a prospective clinical study. A sample was collected 2 h after exemestane dosing at a 1- or 3-month study visit to measure drug concentration. The primary hypothesis was that patients carrying the low-activity CYP3A4*22 (rs35599367) single-nucleotide polymorphism (SNP) would have greater exemestane concentration. Additional SNPs in genes relevant to exemestane metabolism (CYP1A1/2, CYP1B1, CYP3A4, CYP4A11, AKR1C3/4, AKR7A2) were screened in secondary analyses and adjusted for clinical covariates. CYP3A4*22 was associated with a 54% greater exemestane concentration (P<0.01). Concentration was greater in patients who reported White race, had elevated aminotransferases, renal insufficiency, lower body mass index and had not received chemotherapy (all P<0.05), and CYP3A4*22 maintained significance after adjustment for covariates (P<0.01). These genetic and clinical predictors of exemestane concentration may be useful for treatment individualization in patients with breast cancer.


Asunto(s)
Androstadienos/sangre , Antineoplásicos/sangre , Neoplasias de la Mama/genética , Citocromo P-450 CYP3A/genética , Variantes Farmacogenómicas , Polimorfismo de Nucleótido Simple , Androstadienos/administración & dosificación , Androstadienos/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Femenino , Técnicas de Genotipaje , Humanos , Persona de Mediana Edad , Pruebas de Farmacogenómica , Posmenopausia , Medicina de Precisión , Valor Predictivo de las Pruebas
3.
Pharmacogenomics J ; 14(2): 151-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23629159

RESUMEN

Efavirenz increases CYP2C19- and CYP3A-mediated omeprazole metabolism. We hypothesized that CYP2C19 and CYP2B6 genetic polymorphisms influence the extent of induction of omeprazole metabolism by efavirenz. Healthy subjects (n=57) were administered a single 20 mg oral dose of omeprazole on two occasions: with a single 600 mg efavirenz dose; and after a 17-day treatment with efavirenz (600 mg per day). DNA was genotyped for CYP2C19*2, *3 and *17 alleles and CYP2B6*6, *4 and *9 alleles using Taqman assays. Omeprazole, its enantiomers and metabolites were measured by liquid chromatography/tandem mass spectrometry. Our results showed that efavirenz increased omeprazole clearances in all CYP2C19 genotypes in non-stereoselective manner, but the magnitude of induction was genotype dependent. Metabolic ratios of 5-hydroxylation of omeprazole were reduced in extensive and intermediate metabolizers of CYP2C19 (P<0.05). No significant associations were observed between CYP2B6 genotypes and induction by efavirenz on omeprazole metabolism. Our data indicate how interplays between drug interactions and CYP2C19 genetic variations may influence systemic exposure of CYP2C19 substrates.


Asunto(s)
Benzoxazinas/administración & dosificación , Citocromo P-450 CYP2C19/genética , Interacciones Farmacológicas/genética , Omeprazol/administración & dosificación , Adulto , Alquinos , Ciclopropanos , Citocromo P-450 CYP3A/genética , Estudios de Asociación Genética , Genotipo , Humanos , Hidroxilación , Masculino , Persona de Mediana Edad , Polimorfismo Genético
4.
Br J Cancer ; 109(9): 2331-9, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24084768

RESUMEN

BACKGROUND: Change in breast density may predict outcome of women receiving adjuvant hormone therapy for breast cancer. We performed a prospective clinical trial to evaluate the impact of inherited variants in genes involved in oestrogen metabolism and signalling on change in mammographic percent density (MPD) with aromatase inhibitor (AI) therapy. METHODS: Postmenopausal women with breast cancer who were initiating adjuvant AI therapy were enrolled onto a multicentre, randomised clinical trial of exemestane vs letrozole, designed to identify associations between AI-induced change in MPD and single-nucleotide polymorphisms in candidate genes. Subjects underwent unilateral craniocaudal mammography before and following 24 months of treatment. RESULTS: Of the 503 enrolled subjects, 259 had both paired mammograms at baseline and following 24 months of treatment and evaluable DNA. We observed a statistically significant decrease in mean MPD from 17.1 to 15.1% (P<0.001), more pronounced in women with baseline MPD ≥20%. No AI-specific difference in change in MPD was identified. No significant associations between change in MPD and inherited genetic variants were observed. CONCLUSION: Subjects with higher baseline MPD had a greater average decrease in MPD with AI therapy. There does not appear to be a substantial effect of inherited variants in biologically selected candidate genes.


Asunto(s)
Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Mama/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Androstadienos/uso terapéutico , Aromatasa/genética , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quimioterapia Adyuvante/métodos , Estrógenos/metabolismo , Femenino , Humanos , Letrozol , Mamografía/métodos , Persona de Mediana Edad , Nitrilos/uso terapéutico , Polimorfismo de Nucleótido Simple , Posmenopausia/efectos de los fármacos , Posmenopausia/genética , Posmenopausia/metabolismo , Estudios Prospectivos , Triazoles/uso terapéutico
5.
Ann Oncol ; 24(7): 1867-1873, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23508821

RESUMEN

BACKGROUND: Beyond estrogen receptor (ER), there are no validated predictors for tamoxifen (TAM) efficacy and toxicity. We utilized a genome-wide cell-based model to comprehensively evaluate genetic variants for their contribution to cellular sensitivity to TAM. DESIGN: Our discovery model incorporates multidimensional datasets, including genome-wide genotype, gene expression, and endoxifen-induced cellular growth inhibition in the International HapMap lymphoblastoid cell lines (LCLs). Genome-wide findings were further evaluated in NCI60 cancer cell lines. Gene knock-down experiments were performed in four breast cancer cell lines. Genetic variants identified in the cell-based model were examined in 245 Caucasian breast cancer patients who underwent TAM treatment. RESULTS: We identified seven novel single-nucleotide polymorphisms (SNPs) associated with endoxifen sensitivity through the expression of 10 genes using the genome-wide integrative analysis. All 10 genes identified in LCLs were associated with TAM sensitivity in NCI60 cancer cell lines, including USP7. USP7 knock-down resulted in increasing resistance to TAM in four breast cancer cell lines tested, which is consistent with the finding in LCLs and in the NCI60 cells. Furthermore, we identified SNPs that were associated with TAM-induced toxicities in breast cancer patients, after adjusting for other clinical factors. CONCLUSION: Our work demonstrates the utility of a cell-based model in genome-wide identification of pharmacogenomic markers.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Resistencia a Antineoplásicos/genética , Polimorfismo de Nucleótido Simple , Tamoxifeno/análogos & derivados , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral/efectos de los fármacos , Ensayos Clínicos como Asunto , Ensayos de Selección de Medicamentos Antitumorales , Receptor alfa de Estrógeno , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Humanos , ARN Interferente Pequeño/genética , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7
6.
Br J Cancer ; 102(2): 294-300, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-19953095

RESUMEN

BACKGROUND: Tamoxifen, a selective oestrogen receptor (ER) modulator, increases bone mineral density (BMD) in postmenopausal women and decreases BMD in premenopausal women. We hypothesised that inherited variants in candidate genes involved in oestrogen signalling and tamoxifen metabolism might be associated with tamoxifen effects in bone. METHODS: A total of 297 women who were initiating tamoxifen therapy were enrolled in a prospective multicentre clinical trial. Lumbar spine and total hip BMD values were measured using dual-energy X-ray absorptiometry (DXA) at baseline and after 12 months of tamoxifen therapy. Single-nucleotide polymorphisms (SNPs) in ESR1, ESR2, and CYP2D6 were tested for associations in the context of menopausal status and previous chemotherapy, with a mean percentage change in BMD over 12 months. RESULTS: The percentage increase in BMD was greater in postmenopausal women and in those patients who had been treated with chemotherapy. No significant associations between tested SNPs and either baseline BMD or change in BMD with 1 year of tamoxifen therapy were detected. CONCLUSION: The evaluated SNPs in ESR and CYP2D6 do not seem to influence BMD in tamoxifen-treated subjects.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Densidad Ósea/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Tamoxifeno/farmacología , Absorciometría de Fotón , Adulto , Citocromo P-450 CYP2D6/genética , Receptor beta de Estrógeno/genética , Femenino , Humanos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Estudios Prospectivos , Sistema de Registros
7.
Ann Oncol ; 20(12): 1971-6, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19605506

RESUMEN

BACKGROUND: No standard therapy exists for post-docetaxel castrate-resistant prostate cancer (CRPC) patients. This trial aimed to determine the safety and efficacy of pemetrexed in post-docetaxel CRPC patients. MATERIALS AND METHODS: CRPC patients with progression after docetaxel (Taxotere) therapy received pemetrexed (500 mg/m2) i.v. every 3 weeks. The primary end point was prostate-specific antigen (PSA) response. A pharmacogenetic analysis of the reduced folate carrier-1 gene (RFC1) G80A polymorphism was also carried out. RESULTS: Forty-nine patients were enrolled: median age 68 years, median baseline PSA 72 ng/ml, and median Karnofsky performance status of 90. Grade 3 or 4 toxicity occurred in 20 (43%) and four patients (8%), respectively. Confirmed >50% PSA decline occurred in four patients (8%), stable PSA lasting at least 12 weeks in 10 patients (20%). A significant relationship was observed between time from prior docetaxel therapy and overall survival. Pharmacogenetic analyses of RFC1 G80A genotype frequencies showed no relationship between genotypes and clinical efficacy. CONCLUSIONS: Pemetrexed treatment of CRPC patients after docetaxel therapy was associated with only modest clinical activity. Further investigation of pemetrexed as a single agent in a nonenriched CRPC population is unlikely to add significant clinical benefit over that seen with traditional second-line chemotherapy agents such as mitoxantrone.


Asunto(s)
Antineoplásicos/uso terapéutico , Glutamatos/uso terapéutico , Guanina/análogos & derivados , Orquiectomía , Neoplasias de la Próstata/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Glutamatos/efectos adversos , Guanina/efectos adversos , Guanina/uso terapéutico , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Persona de Mediana Edad , Mutación , Metástasis de la Neoplasia , Pemetrexed , Farmacogenética , Polimorfismo Genético , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Análisis de Supervivencia
8.
Pharmacogenomics J ; 9(4): 258-64, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19421167

RESUMEN

The selective estrogen receptor modulator tamoxifen is routinely used for treatment and prevention of estrogen-receptor-positive breast cancer. Studies of tamoxifen adherence suggest that over half of patients discontinue treatment before the recommended 5 years. We hypothesized that polymorphisms in CYP2D6, the enzyme responsible for tamoxifen activation, predict for tamoxifen discontinuation. Tamoxifen-treated women (n=297) were genotyped for CYP2D6 variants and assigned a 'score' based on predicted allele activities from 0 (no activity) to 2 (high activity). Correlation between CYP2D6 score and discontinuation rates at 4 months was tested. We observed a strong nonlinear correlation between higher CYP2D6 score and increased rates of discontinuation (r(2)=0.935, P=0.018). These data suggest that presence of active CYP2D6 alleles may predict for higher likelihood of tamoxifen discontinuation. Therefore, patients who may be most likely to benefit from tamoxifen may paradoxically be most likely to discontinue treatment prematurely.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Citocromo P-450 CYP2D6/genética , Cooperación del Paciente , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Tamoxifeno/uso terapéutico , Citocromo P-450 CYP2D6/metabolismo , Femenino , Humanos , Estudios Prospectivos , Tamoxifeno/efectos adversos , Tamoxifeno/metabolismo
9.
Clin Pharmacol Ther ; 81(3): 328-45, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17339863

RESUMEN

The NIH Pharmacogenetics Research Network (PGRN) is a collaborative group of investigators with a wide range of research interests, but all attempting to correlate drug response with genetic variation. Several research groups concentrate on drugs used to treat specific medical disorders (asthma, depression, cardiovascular disease, addiction of nicotine, and cancer), whereas others are focused on specific groups of proteins that interact with drugs (membrane transporters and phase II drug-metabolizing enzymes). The diverse scientific information is stored and annotated in a publicly accessible knowledge base, the Pharmacogenetics and Pharmacogenomics Knowledge base (PharmGKB). This report highlights selected achievements and scientific approaches as well as hypotheses about future directions of each of the groups within the PGRN. Seven major topics are included: informatics (PharmGKB), cardiovascular, pulmonary, addiction, cancer, transport, and metabolism.


Asunto(s)
Quimioterapia , Farmacogenética , Polimorfismo de Nucleótido Simple , Animales , Fármacos Cardiovasculares/farmacología , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/genética , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/genética , Humanos , Informática , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Preparaciones Farmacéuticas/metabolismo , Inhibidores de Agregación Plaquetaria/uso terapéutico , Trastornos Relacionados con Sustancias/genética , Trastornos Relacionados con Sustancias/rehabilitación
10.
Cancer Res ; 57(16): 3486-93, 1997 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9270017

RESUMEN

Acquired resistance to antiestrogens is a major problem in the clinical management of initially endocrine responsive metastatic breast cancer. We have shown previously that estrogen-independent and -responsive MCF7/LCC1 human breast cancer cells selected for resistance to the triphenylethylene tamoxifen produce a variant (MCF7/LCC2) that retains sensitivity to the steroidal antiestrogen ICI 182,780 (N. Brunner et al., Cancer Res., 53: 3229-3232, 1993). We have now applied stepwise selections in vitro from 10 pM to 1 microM ICI 182,780 against MCF7/LCC1 and obtained a stable ICI 182,780-resistant variant designated MCF7/LCC9. In contrast to 4-hydroxytamoxifen-selected MCF7/LCC2 cells, MCF7/LCC9 cells exhibit full cross-resistance to tamoxifen, despite never having been exposed to this drug. Significantly, tamoxifen cross-resistance arose early in the selection, appearing following selection against only 0.1 nM ICI 182,780. Although limited resistance to ICI 182,780 also was observed, full ICI 182,780 resistance was not detected until the selective pressure increased to 100 nM ICI 182,780. Cross-resistance to tamoxifen persisted throughout these additional selections. Despite their antiestrogen cross-resistance, MCF7/LCC9 cells retain a level of estrogen receptor expression comparable to that of their parental MCF7/LCC1 cells. Whereas MCF7/LCC1 cells retain an estrogen-inducible expression of progesterone receptors, MCF7/LCC9 cells exhibit an up-regulated expression of both progesterone receptor mRNA and protein that is no longer estrogen responsive. Estrogen-independent and -responsive components of the MCF7/LCC9 phenotype are apparent in vivo. These cells form slowly growing tumors in ovariectomized athymic nude mice but respond mitogenically upon estrogenic supplementation. The in vivo growth of MCF7/LCC9 tumors is not affected by treatment with ICI 182,780. Although there is some evidence of tamoxifen stimulation of tumor growth, this did not reach statistical significance. If this pattern of cross-resistance occurs in some breast cancer patients, administering triphenylethylene antiestrogens as a first-line therapy with a cross-over to steroidal compounds upon recurrence may be advantageous.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama , Estradiol/análogos & derivados , Antagonistas de Estrógenos/farmacología , Tamoxifeno/farmacología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Estradiol/farmacología , Femenino , Fulvestrant , Marcadores Genéticos , Humanos , Ratones , Ratones Desnudos , Fenotipo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología
11.
Clin Pharmacol Ther ; 100(1): 63-6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26850569

RESUMEN

Hospital systems increasingly utilize pharmacogenomic testing to inform clinical prescribing. Successful implementation efforts have been modeled at many academic centers. In contrast, this report provides insights into the formation of a pharmacogenomics consultation service at a safety-net hospital, which predominantly serves low-income, uninsured, and vulnerable populations. The report describes the INdiana GENomics Implementation: an Opportunity for the UnderServed (INGENIOUS) trial and addresses concerns of adjudication, credentialing, and funding.


Asunto(s)
Farmacogenética/organización & administración , Proveedores de Redes de Seguridad/organización & administración , Poblaciones Vulnerables , Centros Médicos Académicos/organización & administración , Humanos , Pacientes no Asegurados , Pobreza
12.
Clin Pharmacol Ther ; 98(2): 205-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25968989

RESUMEN

Developmental changes in the liver can significantly impact drug disposition. Due to the emergence of microRNAs (miRNAs) as important regulators of drug disposition gene expression, we studied age-dependent changes in miRNA expression. Expression of 533 miRNAs was measured in 90 human liver tissues (fetal, pediatric [1-17 years], and adult [28-80 years]; n = 30 each). In all, 114 miRNAs were upregulated and 72 were downregulated from fetal to pediatric, and 2 and 3, respectively, from pediatric to adult. Among the developmentally changing miRNAs, 99 miRNA-mRNA interactions were predicted or experimentally validated (e.g., hsa-miR-125b-5p-CYP1A1; hsa-miR-34a-5p-HNF4A). In human liver samples (n = 10 each), analyzed by RNA-sequencing, significant negative correlations were observed between the expression of >1,000 miRNAs and mRNAs of drug disposition and regulatory genes. Our data suggest a mechanism for the marked changes in hepatic gene expression between the fetal and pediatric developmental periods, and support a role for these age-dependent miRNAs in regulating drug disposition.


Asunto(s)
Envejecimiento/genética , Hígado/metabolismo , MicroARNs/genética , Farmacogenética , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Biotransformación/genética , Niño , Preescolar , Análisis por Conglomerados , Biología Computacional , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Lactante , MicroARNs/metabolismo , Persona de Mediana Edad
13.
Clin Pharmacol Ther ; 98(2): 127-34, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25974703

RESUMEN

Selective serotonin reuptake inhibitors (SSRIs) are primary treatment options for major depressive and anxiety disorders. CYP2D6 and CYP2C19 polymorphisms can influence the metabolism of SSRIs, thereby affecting drug efficacy and safety. We summarize evidence from the published literature supporting these associations and provide dosing recommendations for fluvoxamine, paroxetine, citalopram, escitalopram, and sertraline based on CYP2D6 and/or CYP2C19 genotype (updates at www.pharmgkb.org).


Asunto(s)
Citocromo P-450 CYP2C19/genética , Citocromo P-450 CYP2D6/genética , Cálculo de Dosificación de Drogas , Farmacogenética/normas , Polimorfismo Genético , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Biotransformación , Citocromo P-450 CYP2C19/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Genotipo , Humanos , Seguridad del Paciente , Fenotipo , Medición de Riesgo , Factores de Riesgo , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacocinética
14.
Endocrinology ; 142(4): 1497-505, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11250930

RESUMEN

Many human breast tumors are driven by high intratumor concentrations of 17beta-estradiol that appear to be locally synthesized. The role of aromatase is well established, but the possible contribution of the steroid sulfatase (STS), which liberates estrogens from their biologically inactive sulfates, has been inadequately assessed and remains unclear. To evaluate the role of STS further, we transduced estrogen-dependent MCF-7 human breast cancer cells with a retroviral vector directing the constitutive expression of the human STS gene. Gene integration was confirmed by Southern hybridization, production of the appropriately sized messenger RNA by Northern hybridization, and expression of functional protein by metabolism of [(3)H]estrone sulfate to [(3)H]estrone. Maximum velocity estimates of estrone formation are 64.2 pmol estrone/mg protein.h in STS-transduced cells (STS Clone 20), levels comparable to those seen in some human breast tumors. Lower levels of endogenous activity are seen in MCF-7 cells (13.0 pmol estrone/mg protein.h) and in cells transduced with vector lacking the STS gene (Vector 3 cells; 12.0 pmol estrone/mg protein.h). 17beta-Estradiol sulfate induces expression of the progesterone receptor messenger RNA only in STS Clone 20 cells, whereas estrone sulfate produces the greatest stimulation of anchorage-independent growth in these cells. STS Clone 20 cells retain responsiveness to antiestrogens, which block the ability of estrogen sulfate to increase the proportion of cells in both the S and G(2)/M phases of the cell cycle. Consistent with these in vitro observations, only STS Clone 20 cells exhibit a significant increase in the proportion of proliferating tumors in nude ovariectomized mice supplemented with 17beta-estradiol sulfate. The primary activity in vivo appears to be from intratumor STS, rather than hepatic STS. Surprisingly, 17beta-estradiol sulfate appears more effective than 17beta-estradiol when both are administered at comparable concentrations. This effect, which is seen only in STS Clone 20 cells, may reflect differences in the cellular pharmacology of exogenous estrogens compared with those released by the activity of intracellular STS. These studies directly demonstrate that intratumor STS activity can support estrogen-dependent tumorigenicity in an experimental model and may contribute to the promotion of human breast tumors.


Asunto(s)
Arilsulfatasas/biosíntesis , Arilsulfatasas/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Animales , Northern Blotting , Southern Blotting , Neoplasias de la Mama/patología , Ciclo Celular/fisiología , Estradiol/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Ensayos de Protección de Nucleasas , Transducción de Señal/efectos de los fármacos , Esteril-Sulfatasa , Fracciones Subcelulares/metabolismo , Células Tumorales Cultivadas
15.
Cancer Epidemiol Biomarkers Prev ; 7(12): 1109-15, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9865429

RESUMEN

Nucleophosmin (NPM) is an estrogen-regulated nucleolar phosphoprotein; a substrate for phosphorylation by p34cdc2 kinase, protein kinase C, and casein kinase II; and a repressor of the transcriptional regulating activities of the YY1 and IFN regulatory factor-1 transcription factors. We have completed a pilot study to determine whether autoantibodies to NPM are present in breast cancer patients and explored the ability of these autoantibodies to predict recurrence in breast cancer patients. One hundred breast cancer patients were studied: 50 who recurred, and 50 matched for age and length of follow-up but who did not recur. Patients' sera were collected at the times of diagnosis (T1), six months before recurrence (T2), and at recurrence (T3). Recurrent and nonrecurrent patients did not differ in autoantibody levels at the times of diagnosis or recurrence. However, antiNPM autoantibody levels increase significantly between diagnosis and six months before recurrence in recurrent patients, whereas no change occurs over the comparable time period in nonrecurrent patients (repeated measures ANOVA; P = 0.041). At recurrence, the levels return to those seen at diagnosis. The greater the change in levels between T1 and T2, the greater the risk of recurrence within the next 6 months (conditional logistic regression: increase in risk for highest versus lowest tertile of change from T1 to T2; odds ratio, 3.25; 95% confidence interval, 1.04-10.18; P = 0.043). Consistent with the estrogenic/antiestrogenic regulation of the antigen in breast cancer cells, the levels of antiNPM autoantibodies are decreased 6 months before recurrence in patients treated with the antiestrogen tamoxifen (P = 0.012). The association between antiNPM levels and recurrence remained after adjustment for confounding factors. Further study of antiNPM autoantibody levels as a new and simple, intermediate serum biomarker for predicting both the timing of recurrence and monitoring response to endocrine manipulations in breast cancer patients is warranted.


Asunto(s)
Autoanticuerpos/sangre , Neoplasias de la Mama/inmunología , Recurrencia Local de Neoplasia/inmunología , Neoplasias Hormono-Dependientes/inmunología , Proteínas Nucleares/inmunología , Fosfoproteínas/inmunología , Adulto , Análisis de Varianza , Biomarcadores de Tumor/sangre , Estudios de Casos y Controles , Estrógenos , Femenino , Humanos , Persona de Mediana Edad , Nucleofosmina , Oportunidad Relativa , Proyectos Piloto
16.
J Endocrinol ; 128(2): 219-28, 1991 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1706405

RESUMEN

In vitro, insulin-like growth factor-I (IGF-I) promotes both growth and development of bovine mammary tissue. In vivo, the effects of IGF-I may encompass endocrine, paracrine or autocrine mediation. We addressed the possibility of paracrine/autocrine effects of IGF-I in the mammary gland by examining the in-vitro secretion of IGF-I and IGF-binding proteins (IGFBPs) from bovine mammary tissue. Bovine mammary explants from pregnant non-lactating and lactating non-pregnant animals were found to synthesize and secrete IGF-I and IGFBPs. Mammary acini cultures, representative of mammary secretory epithelia, secreted both IGF-I and IGFBP, but synthesized only IGFBP. Concentrations of IGF-I in conditioned media from explants were 1.54 and 0.72 fmol/micrograms DNA for pregnant and lactating animals respectively. Concentrations of IGFBPs in conditioned media from explants were similar for both physiological states at 2529 pmol 125I-labelled IGF-I bound/micrograms DNA. Ligand/Western blotting procedures identified four IGFBPs of 29, 33, 37 and 44 kDa for acini cultures and five IGFBPs of 28, 31, 36, 44 and 46 kDa for explant cultures. Similar affinities for IGF-I and IGF-II were shown by IGFBP, using 125I-labelled recombinant human IGF-I as the competing ligand (median effective dose (ED50) of 0.085 pmol). When 125I-labelled bovine IGF-II was used as the ligand, only bovine IGF-II (ED50 of 0.25 pmol) inhibited binding. The addition of prolactin, insulin and cortisol, with or without GH, did not affect secretion of either IGF-I or IGFBP. This report describes the ability of normal mammary tissue to synthesize and secrete IGF-I and IGFBPs.


Asunto(s)
Proteínas Portadoras/fisiología , Bovinos/fisiología , Glándulas Mamarias Animales/fisiología , Preñez/fisiología , Somatomedinas/fisiología , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/metabolismo , Técnicas de Cultivo , Femenino , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lactancia/fisiología , Glándulas Mamarias Animales/metabolismo , Embarazo , Somatomedinas/biosíntesis , Somatomedinas/metabolismo
17.
J Endocrinol ; 131(1): 127-33, 1991 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1720804

RESUMEN

The bovine mammary gland accumulates large quantities of insulin-like growth factors (IGFs) and IGF-binding proteins (IGFBPs) during late gestation which are secreted at parturition. The present study was conducted to determine the changes in the profiles of IGFBPs secreted by the mammary gland and in blood during late gestation and early lactation in dairy cows. Ligand blotting of serum and mammary secretions showed that IGFBPs of Mr 25,000, 30,000, 34,000, 42,000, 46,000 and greater than 200,000 were present in both fluids. The binding activity of the 42-46,000 Mr IGFBP predominated in prepartum mammary secretions and colostrum but was reduced postpartum. The binding activities of the 30,000 and 34,000 Mr IGFBPs, relative to other IGFBPs, were increased postpartum. Concentrations of IGF-I and IGF-II in mammary secretions declined from 347.1 and 181.1 nmol/litre 1 week prepartum to 0.7 and 0.3 nmol/litre 1.5 weeks postpartum. The volume of mammary secretions obtained was 0.109 litre and 6.690 litres at 1 week prepartum and 1.5 weeks postpartum respectively. In prepartum serum, the greatest binding activity was at Mr 42-46,000. The activity at this Mr decreased at parturition but was restored postpartum. The binding activities of the 30,000 and 34,000 Mr IGFBPs were increased around parturition. The 25,000 Mr IGFBP had minor activity during all periods. IGF-I concentrations decreased from 10.6 nmol/litres 1 week prepartum to 4.7 nmol/litres 1.5 weeks postpartum but IGF-II concentrations remained constant. In conclusion, IGFBP activity secreted by the mammary gland shifts from primarily Mr 42-46,000 prepartum to Mr 30,000 postpartum.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Proteínas Portadoras/metabolismo , Bovinos/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Trabajo de Parto/metabolismo , Leche/metabolismo , Preñez/metabolismo , Animales , Autorradiografía , Proteínas Portadoras/sangre , Calostro/metabolismo , Femenino , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina , Factor II del Crecimiento Similar a la Insulina/metabolismo , Glándulas Mamarias Animales/metabolismo , Embarazo
18.
J Steroid Biochem Mol Biol ; 67(5-6): 391-402, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10030688

RESUMEN

We have used two-dimensional gel electrophoresis to identify proteins associated with estrogen-induced proliferation in MCF-7 breast cancer cells and their progression to estrogen-independent proliferation. We compared the total cellular proteins from MCF-7 cells and an estrogen independent derivative of the MCF-7 cells MCF-7/LCC1 (Brünner et al. Cancer Research 1993, 53, 283-290), each grown with and without estradiol. These comparisons reveal seven estrogen-regulated proteins. Three of these proteins (HI-1: 36 kDa/pI 4.5, HI-10: 40 kDa/pI 5.5 and HI-19: 62 kDa/pI 5.0) exhibit a 'progression-like' pattern, being induced by estradiol in MCF-7 cells and constitutively present/upregulated in the MCF-7/LCC1 growing without estradiol. HI-11 (65 kDa/pI 5.5) is strongly induced by estradiol in MCF-7 cells but constitutively downregulated and unresponsive to estradiol in the MCF-7/LCC1 cells. Two proteins exhibit a suppressor pattern and are downregulated by estradiol in the estrogen-dependent MCF-7 cells (HI-3: 44 kDa/pI 4.4 and HI-4: 56 kDa/ pI 5.2) and present in MCF-7/LCC1 cells growing without estradiol at levels comparable to that seen in estrogen-treated MCF-7 cells. One protein (HI-9: 68 kDa/pI 5.5) exhibits a marked estrogen regulated pI shift, rather than changes in abundance. We purified and sequenced the HI-10 protein, which we identified as the nucleolar protein, nucleophosmin (NPM). One- and two-dimensional Western blot analyses of MCF-7/LCC1 cell lysates confirmed that HI-10 is immunoreactive with an antinucleophosmin antibody. Western blotting also confirmed the estrogenic regulation of NPM seen in the initial two-dimensional gel electrophoresis studies. Thus, NPM is induced by estradiol in the MCF-7 cells and upregulated in the MCF-7/LCC1 cells growing without estrogen, clearly associating its expression with an acquired estrogen-independent phenotype. NPM has several potentially important roles in regulating cell function and signaling. It is a substrate for phosphorylation by p34cdc2 kinase, protein kinase C and nuclear kinase II, and a repressor of the transcriptional regulating activities of both the IRF-1 tumor suppressor protein and the YY1 transcription factor. Studies are currently underway to determine which of these NPM functions may be involved in the hormonal progression of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Nucleares/genética , Secuencia de Aminoácidos , Núcleo Celular/metabolismo , Electroforesis en Gel Bidimensional , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/aislamiento & purificación , Nucleofosmina , Fragmentos de Péptidos/química , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/aislamiento & purificación , Células Tumorales Cultivadas
19.
J Steroid Biochem Mol Biol ; 76(1-5): 71-84, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11384865

RESUMEN

Endocrine therapy is effective in approximately one-third of all breast cancers and up to 80% of tumors that express both estrogen and progesterone receptors. Despite the low toxicity, good overall response rates, and additional benefits associated with its partial agonist activity, most Tamoxifen-responsive breast cancers acquire resistance. The development of new antiestrogens, both steroidal and non-steroidal, provides the opportunity for the development of non-cross-resistant therapies and the identification of additional mechanisms of action and resistance. Drug-specific pharmacologic mechanisms may confer a resistance phenotype, reflecting the complexities of both tumor biology/pharmacology and the molecular endocrinology of steroid hormone action. However, since all antiestrogens will be effective only in cells that express estrogen receptors (ER), many mechanisms will likely be directly related to ER expression and signaling. For example, loss of ER expression/function is likely to confer a cross-resistance phenotype across all structural classes of antiestrogens. Altered expression of ERalpha and ERbeta, and/or signaling from transcription complexes driven by these receptors, may produce drug-specific resistance phenotypes. We have begun to study the possible changes in gene expression that may occur as cells acquire resistance to steroidal and non-steroidal antiestrogens. Our preliminary studies implicate the altered expression of several estrogen-regulated genes. However, resistance to antiestrogens is likely to be a multigene phenomenon, involving a network of interrelated signaling pathways. The way in which this network is adapted by cells may vary among tumors, consistent with the existence of a highly plastic and adaptable genotype within breast cancer cells.


Asunto(s)
Moduladores de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , División Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Moduladores de los Receptores de Estrógeno/uso terapéutico , Sustancias de Crecimiento/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/enzimología , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Estrés Oxidativo , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/fisiología , Transducción de Señal , Tamoxifeno/uso terapéutico
20.
J Med Food ; 2(3-4): 143-9, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-19281366

RESUMEN

Estrogens primarily function through the activation of their receptors, which subsequently function as nuclear transcription factors. There are two estrogen receptor (ER) genes, now designated ERa (the classic ER gene) and ER/3. The key consequence of the activation of either gene product is the regulation of gene transcription. The extent and nature of transcription appear to be regulated by a series of coregulator proteins. One of the most sensitive assays for detection of potential estrogenic activity is measurement of the ability of a test compound to influence the transcription of reporter genes. In this regard, many investigators use promoter-reporter constructs. To assess putative estrogenic activity, an estrogen-responsive promoter is generally placed upstream of a reporter gene and transiently transfected into a target cell. When exposed to an estrogenic compound, expression of the reporter gene would normally be induced. We briefly discuss several issues pertinent to the use of these assays and the interpretation of resulting data, including estrogen-responsive, promoter-reporter constructs, reporter genes and measurements of activity, choice of target cell or cell line, transient introduction of promoter-reporter constructs into cells, basic statistical approaches to data analysis, and definitions of agonist, partial agonist, and antagonist.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda