Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Support Care Cancer ; 32(7): 407, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38833106

RESUMEN

OBJECTIVE: Poor adherence to oral chemotherapy adversely impacts clinical outcomes and escalates overall healthcare costs. Despite barriers to medication adherence, a significant gap remains in assessing adherence to oral chemotherapy among multiple myeloma (MM) patients with lower socioeconomic status. Hence, our study aims to evaluate immunomodulator adherence in MM patients at a county hospital, primarily serving underrepresented and indigent individuals with low socioeconomic status across the greater Houston area. METHODS: Inclusion criteria composed of patients diagnosed with MM, aged at least 18 years, and treated with lenalidomide or pomalidomide-two widely used immunomodulators-for a minimum of 2 months or having two or more records of dispensation between May 2019 and May 2021. Adherence was gauged using an adjusted version of the medication possession ratio (MPR). RESULTS: Sixty-two patients were enrolled, yielding a mean MPR value of 88% (SD, ± 18.9). Of these, 43 patients (69.3%) demonstrated adherence with an MPR of ≥ 0.90. A significant difference was found in treatment duration between the adherent (mean 8.8 months; SD, ± 7.2) and non-adherent (mean 13.4 months; SD, ± 7.9) groups (p = 0.027). Notably, race/ethnicity demonstrated a significant difference (p = 0.048), driven by disparities in African American and Hispanic representation across adherence levels. CONCLUSION: In summary, our findings highlight race and treatment duration to be predictors of immunomodulator adherence among MM patients with lower socioeconomic status. Further research is imperative to devise and test innovative interventions aimed at enhancing medication adherence, thereby contributing to improved survival and healthcare quality in this population.


Asunto(s)
Lenalidomida , Cumplimiento de la Medicación , Mieloma Múltiple , Clase Social , Talidomida , Humanos , Mieloma Múltiple/tratamiento farmacológico , Masculino , Estudios Retrospectivos , Cumplimiento de la Medicación/estadística & datos numéricos , Femenino , Persona de Mediana Edad , Anciano , Talidomida/uso terapéutico , Talidomida/análogos & derivados , Talidomida/administración & dosificación , Lenalidomida/administración & dosificación , Lenalidomida/uso terapéutico , Factores Inmunológicos/uso terapéutico , Factores Inmunológicos/administración & dosificación , Agentes Inmunomoduladores/uso terapéutico , Agentes Inmunomoduladores/administración & dosificación , Agentes Inmunomoduladores/farmacología , Texas , Anciano de 80 o más Años , Adulto
2.
Breast Cancer Res Treat ; 195(3): 421-430, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35969285

RESUMEN

PURPOSE: Metformin has demonstrated a chemoprotective effect in breast cancer but there is limited evidence on the effect of cumulative exposure to metformin and the risk of hormone receptor-positive and human epidermal growth factor receptor 2-negative (HR + /HER2-) breast cancer. This study assessed this risk with dose and intensity of metformin in postmenopausal women with type-2 diabetes mellitus (T2DM). METHODS: This nested case-control study used the Surveillance, Epidemiology, and End Results-Medicare data (2008-2015). Cohort entry was the date of incident T2DM diagnosis. Cases were those diagnosed with HR + /HER2- breast cancer (event date) as their first/only cancer. Non-cancer T2DM controls were matched using variable-ratio-matching. Cumulative dose and average intensity of metformin were measured during the 1-year lookback period. Dose(mg) was categorized as: (1)0, (2)0-30,000, (3)30,001-136,000, (4)136,001-293,000, and (5) > 293,000, and intensity(mg/day) as: 0, 1-500, and > 500. Covariates were conceptualized using the Andersen Behavioral Model. Conditional logistic regression was used to assess the risk of HR + /HER2- breast cancer with metformin-use. RESULTS: There were 690 cases and 2747 controls. The median duration of T2DM was 1178 days in controls and 1180 days in cases. Higher cumulative dose categories: 4 (adjusted odds ratio(aOR) = 0.72, 95% CI 0.55-0.95,p = 0.02), and 5 (OR = 0.60, 95% CI 0.42-0.85,p < 0.01) had significantly lower odds of HR + /HER2- breast cancer compared to category 0. The highest intensity category of metformin had 39% lower odds of HR + /HER2- breast cancer (OR = 0.61, 95% CI 0.46-0.82,p < 0.01) compared to the 0 mg/day group. CONCLUSIONS: Higher metformin exposure was associated with reduced risk of HR + /HER2- breast cancer, adding to the evidence supporting metformin's chemoprotective effect.


Asunto(s)
Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Metformina , Anciano , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/metabolismo , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/epidemiología , Femenino , Humanos , Medicare , Metformina/uso terapéutico , Posmenopausia , Receptor ErbB-2/metabolismo , Estados Unidos/epidemiología
3.
FASEB J ; 35(7): e21719, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34110646

RESUMEN

While G protein-coupled receptors (GPCRs) are known to be excellent drug targets, the second largest family of adhesion-GPCRs is less explored for their role in health and disease. ADGRF1 (GPR110) is an adhesion-GPCR and has an important function in neurodevelopment and cancer. Despite serving as a poor predictor of survival, ADGRF1's coupling to G proteins and downstream pathways remain unknown in cancer. We evaluated the effects of ADGRF1 overexpression on tumorigenesis and signaling pathways using two human epidermal growth factor receptor-2-positive (HER2+) breast cancer (BC) cell-line models. We also interrogated publicly available clinical datasets to determine the expression of ADGRF1 in various BC subtypes and its impact on BC-specific survival (BCSS) and overall survival (OS) in patients. ADGRF1 overexpression in HER2+ BC cells increased secondary mammosphere formation, soft agar colony formation, and % of Aldefluor-positive tumorigenic population in vitro and promoted tumor growth in vivo. ADGRF1 co-immunoprecipitated with both Gαs and Gαq proteins and increased cAMP and IP1 when overexpressed. However, inhibition of only the Gαs pathway by SQ22536 reversed the pro-tumorigenic effects of ADGRF1 overexpression. RNA-sequencing and RPPA analysis revealed inhibition of cell cycle pathways with ADGRF1 overexpression, suggesting cellular quiescence, as also evidenced by cell cycle arrest at the G0/1 phase and resistance to chemotherapy in HER2+ BC. ADGRF1 was significantly overexpressed in the HER2-enriched BC compared to luminal A and B subtypes and predicted worse BCSS and OS in these patients. Therefore, ADGRF1 represents a novel drug target in HER2+ BC, warranting discovery of novel ADGRF1 antagonists.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas Oncogénicas/genética , Receptor ErbB-2/genética , Receptores Acoplados a Proteínas G/genética , Animales , Neoplasias de la Mama/genética , Carcinogénesis/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Fase G1/genética , Humanos , Ratones , Ratones Desnudos , Fase de Descanso del Ciclo Celular/genética , Transducción de Señal/genética
4.
Breast Cancer Res Treat ; 182(2): 401-409, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32500397

RESUMEN

PURPOSE: Obesity and insulin resistance have been associated with poor prognosis in breast cancer (BC). The present prospective study aimed to investigate the impact of metabolic syndrome (MetS) and its components on early BC (eBC) patients' outcome. METHODS: MetS was defined by the presence of 3 to 5 of the following components: waist circumference > 88 cm, blood pressure ≥ 130/≥ 85 mmHg, serum levels of triglycerides ≥ 150 mg/dL, high density lipoprotein < 50 mg/dL and fasting glucose ≥ 110 mg/dL. Seven hundred and seventeen patients with data on ≥ 4 MetS components at BC diagnosis were enrolled. Study population was divided into two groups: patients with < 3 (non-MetS) vs. ≥ 3 components (MetS). Categorical variables were analyzed by Chi-square test and survival data by log-rank test and Cox proportional hazards regression model. RESULTS: Overall, 544 (75.9%) and 173 (24.1%) women were categorized as non-MetS and MetS, respectively. MetS patients were more likely to be older, postmenopausal, and insulin-resistant compared to non-MetS patients (p < 0.05). In multivariate analysis, MetS patients had a numerically higher risk of relapse [disease-free survival (DFS), hazard ratio (HR) 1.51, p = 0.07] and a significantly higher risk of death compared to non-MetS patients [overall survival (OS), HR 3.01, p < 0.0001; breast cancer-specific survival (BCSS), HR 3.16, p = 0.001]. Additionally, patients with 1 to 2 components of MetS had an increased risk of dying compared to patients with 0 components (OS, HR 4.90, p = 0.01; BCSS, HR 6.07, p = 0.02). CONCLUSIONS: MetS correlated with poor outcome in eBC patients. Among patients without full criteria for MetS diagnosis, the presence of 1 or 2 components of the syndrome may predict for worse survival.


Asunto(s)
Neoplasias de la Mama/mortalidad , Síndrome Metabólico/epidemiología , Factores de Edad , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Comorbilidad , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Síndrome Metabólico/diagnóstico , Persona de Mediana Edad , Estudios Prospectivos , Medición de Riesgo , Factores de Riesgo , Circunferencia de la Cintura
5.
Support Care Cancer ; 28(4): 1809-1816, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31338641

RESUMEN

PURPOSE: The impact of supportive medications on patient-reported outcomes (PROs) has not been systematically evaluated. We describe the supportive medications used by treatment-naïve lung cancer patients and assess their association with PROs from MD Anderson Symptom Inventory (MDASI). METHODS: Treatment-naïve lung cancer patients who completed PROs from MDASI at the initial visit to MD Anderson Cancer Center were included. Medications from the initial visit were abstracted from the electronic medical records system and categorized into therapeutic classes based on U.S. Pharmacopeia v7.0. A chi-square or Mann-Whitney U test was conducted as appropriate. RESULTS: Among 459 patients, ~ 50% took any analgesics and 25% were on opioids. One-third of patients with moderate-severe pain were not on any analgesics. Patients taking opioids had significantly worse median pain scores (6 vs. 0) compared with those not taking any analgesics (p < 0.0001). Higher proportion of patients with moderate-severe pain took opioids compared with those with mild pain (52% vs. 16%, p < 0.0001). Patients on opioids also reported significantly worse scores for five other cancer-specific core symptoms and all six symptoms rating interference with daily life. Only 15% of patients with higher composite score for depression-related symptoms were on antidepressants. However, patients taking antidepressants did not significantly differ in any individual MDASI symptom scores compared with those not on antidepressants (p = 0.4858). CONCLUSIONS: Our results suggest a need for better screening for pain and depression and optimization of pain management in treatment-naïve lung cancer patients since their poor functional status may result in suboptimal cancer therapy.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Cuidados Paliativos/métodos , Medición de Resultados Informados por el Paciente , Medicamentos bajo Prescripción/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Analgésicos Opioides/uso terapéutico , Antidepresivos/uso terapéutico , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/epidemiología , Depresión/tratamiento farmacológico , Depresión/epidemiología , Femenino , Humanos , Neoplasias Pulmonares/epidemiología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/psicología , Masculino , Persona de Mediana Edad , Manejo del Dolor/métodos , Manejo del Dolor/psicología , Manejo del Dolor/estadística & datos numéricos , Cuidados Paliativos/estadística & datos numéricos , Polifarmacia , Medicamentos bajo Prescripción/clasificación , Estudios Retrospectivos , Adulto Joven
6.
BMC Cancer ; 19(1): 220, 2019 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-30871481

RESUMEN

BACKGROUND: Breast cancer patient-derived xenograft (BC-PDX) models represent a continuous and reproducible source of circulating tumor cells (CTCs) for studying their role in tumor biology and metastasis. We have previously shown the utility of BC-PDX models in the study of CTCs by immunohistochemistry (IHC) on serial paraffin sections and manual microscopic identification of cytokeratin-positive cells, a method that is both low-throughput and labor-intensive. We therefore aimed to identify and characterize CTCs from small volume mouse blood samples and examined its practical workflow in a study of BC-PDX mice treated with chemotherapy using an automated imaging platform, the AccuCyte®-CyteFinder® system. METHODS: CTC analysis was conducted using blood from non-tumor bearing SCID/Beige mice spiked with human breast cancer cells, BC-PDX-bearing mice, and BC-PDX mice treated with vehicle or chemotherapeutic agent(s). After red blood cell lysis, nucleated cells were mixed with transfer solution, processed onto microscope slides, and stained by immunofluorescence. The CyteFinder automated scanning microscope was used to identify CTCs, defined as nucleated cells that were human cytokeratin-positive, and mouse CD45-negative. Disaggregated primary BC-PDX tumors and lung metastatic nodules were processed using the same immunostaining protocol. Collective expression of breast cancer cell surface markers (EpCAM, EGFR, and HER2) using a cocktail of target-specific antibodies was assessed. CTCs and disaggregated tumor cells were individually retrieved from slides using the CytePicker® module for sequence analysis of a BC-PDX tumor-specific PIK3CA mutation. RESULTS: The recovery rate of human cancer cells spiked into murine blood was 83 ± 12%. CTC detection was not significantly different from the IHC method. One-third of CTCs did not stain positive for cell surface markers. A PIK3CA T1035A mutation present in a BC-PDX tumor was confirmed in isolated single CTCs and cells from dissociated metastatic nodules after whole genome amplification and sequencing. CTC evaluation could be simply implemented into a preclinical PDX therapeutic study setting with substantial improvements in workflow over the IHC method. CONCLUSIONS: Analysis of small volume blood samples from BC-PDX-bearing mice using the AccuCyte-CyteFinder system allows investigation of the role of CTCs in tumor biology and metastasis independent of surface marker expression.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/genética , Células Neoplásicas Circulantes/metabolismo , Análisis de la Célula Individual/métodos , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Separación Celular , Fosfatidilinositol 3-Quinasa Clase I/sangre , Femenino , Humanos , Queratinas/sangre , Antígenos Comunes de Leucocito/sangre , Ratones , Ratones SCID , Mutación , Trasplante de Neoplasias , Células Neoplásicas Circulantes/efectos de los fármacos , Análisis de Secuencia de ADN
7.
Ann Pharmacother ; 53(5): 501-509, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30522347

RESUMEN

OBJECTIVE: To review the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of ribociclib (LEE011, Kisqali) in hormone receptor-positive/human epidermal growth factor receptor-2-negative (HR+/HER2-) metastatic breast cancer. DATA SOURCES: A PubMed search was performed using the terms 'Ribociclib', 'Kisqali', and 'LEE011' between May 2018 and November 2018. References of published articles and reviews were also assessed for additional information. STUDY SELECTION AND DATA EXTRACTION: English-language preclinical and clinical studies on the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of ribociclib were evaluated. DATA SYNTHESIS: Ribociclib, a cyclin-dependent kinase (CDK) 4/6 inhibitor, is Food and Drug Administration (FDA) approved in combination with endocrine therapy for treatment of HR+/HER2- advanced or metastatic breast cancer in premenopausal/perimenopausal and postmenopausal women. Three phase III trials have evaluated ribociclib in combination with endocrine therapy, including letrozole, anastrozole, tamoxifen, and fulvestrant. These studies found that ribociclib 600 mg/d, 21 days on, 7 days off, leads to a significantly greater median progression-free survival (PFS), ranging from 8 to 13 months. Ribociclib is well tolerated in elderly patients, maintains health-related quality of life, and significantly reduces pain scores. The dose-limiting toxicities found in phase I studies were neutropenia, thrombocytopenia, and QTc prolongation. Common adverse effects seen in phase III trials include neutropenia, leukopenia, nausea, diarrhea, vomiting, and fatigue. Relevance to Patient Care and Clinical Practice: Literature on the safety and efficacy of ribociclib as well as its place in therapy in comparison to other FDA-approved CDK4/6 inhibitors for breast cancer is discussed. CONCLUSIONS: Ribociclib, when added to endocrine therapy, significantly improves PFS and has manageable toxicity in premenopausal/perimenopausal and postmenopausal women with HR+/HER2- advanced breast cancer.


Asunto(s)
Aminopiridinas/administración & dosificación , Aminopiridinas/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Purinas/administración & dosificación , Purinas/efectos adversos , Adulto , Anciano , Aminopiridinas/química , Aminopiridinas/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Metástasis de la Neoplasia , Premenopausia/efectos de los fármacos , Premenopausia/fisiología , Purinas/química , Purinas/farmacocinética , Calidad de Vida , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo
8.
Ann Pharmacother ; 53(6): 612-620, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30607980

RESUMEN

OBJECTIVE: To review the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of neratinib in human epidermal growth factor receptor (HER2)+ breast cancer (BC). DATA SOURCES: A PubMed search was performed using the term neratinib between September 12, 2018, and November 21, 2018. References of published articles and reviews were also assessed for additional information. STUDY SELECTION AND DATA EXTRACTION: English-language preclinical and clinical studies on the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of neratinib were evaluated. DATA SYNTHESIS: Neratinib, an irreversible inhibitor of HER1, HER2, and HER4, is Food and Drug Administration approved for the extended adjuvant treatment of stage I-III HER2+ BC to follow trastuzumab-based therapy. A phase III study has demonstrated statistically significant improvement in 5-year disease-free survival rate (90.2 vs 87.7; hazard ratio = 0.73, 95% CI = 0.57-0.92, P = 0.0083). Its most common adverse effect is diarrhea, observed in more than 90% of patients. The incidence of grade 3/4 diarrhea (~40%) is reduced by half with loperamide prophylaxis, which is recommended for the first 8 weeks of neratinib therapy. Other common adverse reactions are nausea and fatigue. The patients need to be monitored for liver function tests and drug interactions with acid-reducing agents, CYP3A4 inhibitors/inducers, and P-glycoprotein substrates with narrow therapeutic window. Relevance to Patient Care and Clinical Practice: American Society of Clinical Oncology and National Comprehensive Cancer Network clinical guidelines suggest the use of neratinib for extended adjuvant therapy following 1-year trastuzumab in stage I to III HER2+ BC. Diarrhea remains a clinically significant but manageable adverse event. CONCLUSION: Neratinib significantly improves treatment outcomes and has manageable toxicity in stage I to III HER2+ BC patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quinolinas/uso terapéutico , Adulto , Femenino , Humanos , Persona de Mediana Edad , Quinolinas/farmacología , Resultado del Tratamiento
9.
Proc Natl Acad Sci U S A ; 113(43): E6600-E6609, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27791031

RESUMEN

Forkhead box protein A1 (FOXA1) is a pioneer factor of estrogen receptor α (ER)-chromatin binding and function, yet its aberration in endocrine-resistant (Endo-R) breast cancer is unknown. Here, we report preclinical evidence for a role of FOXA1 in Endo-R breast cancer as well as evidence for its clinical significance. FOXA1 is gene-amplified and/or overexpressed in Endo-R derivatives of several breast cancer cell line models. Induced FOXA1 triggers oncogenic gene signatures and proteomic profiles highly associated with endocrine resistance. Integrated omics data reveal IL8 as one of the most perturbed genes regulated by FOXA1 and ER transcriptional reprogramming in Endo-R cells. IL-8 knockdown inhibits tamoxifen-resistant cell growth and invasion and partially attenuates the effect of overexpressed FOXA1. Our study highlights a role of FOXA1 via IL-8 signaling as a potential therapeutic target in FOXA1-overexpressing ER-positive tumors.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Interleucina-8/genética , Transcriptoma , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/metabolismo , Pronóstico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Análisis de Supervivencia , Tamoxifeno/uso terapéutico
10.
Breast Cancer Res Treat ; 170(2): 279-292, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29574636

RESUMEN

PURPOSE: G protein-coupled receptors (GPCRs) represent the largest family of druggable targets in human genome. Although several GPCRs can cross-talk with the human epidermal growth factor receptors (HERs), the expression and function of most GPCRs remain unknown in HER2+ breast cancer (BC). In this study, we aimed to evaluate gene expression of GPCRs in tumorigenic or anti-HER2 drug-resistant cells and to understand the potential role of candidate GPCRs in HER2+ BC. METHODS: Gene expression of 352 GPCRs was profiled in Aldeflur+ tumorigenic versus Aldeflur- population and anti-HER2 therapy-resistant derivatives versus parental cells of HER2+ BT474 cells. The GPCR candidates were confirmed in 7 additional HER2+ BC cell line models and publicly available patient dataset. Anchorage-dependent and anchorage-independent cell growth, mammosphere formation, and migration/invasion were evaluated upon GPR110 knockdown by siRNA in BT474 and SKBR3 parental and lapatinib+ trastuzumab-resistant (LTR) cells. RESULTS: Adhesion and class A GPCRs were overexpressed in Aldeflur+ and anti-HER2 therapy-resistant population of BT474 cells, respectively. GPR110 was the only GPCR overexpressed in Aldeflur+ and anti-HER2 therapy-resistant population in BT474, SKBR3, HCC1569, MDA-MB-361, AU565, and/or HCC202 cells and in HER2+ BC subtype in patient tumors. Using BT474 and SKBR3 parental and LTR cells, we found that GPR110 knockdown significantly reduced anchorage-dependent/independent cell growth as well as migration/invasion of parental and LTR cells and mammosphere formation in LTR derivatives and not in parental cells. CONCLUSION: Our data suggest a potential role of GPR110 in tumorigenicity and in tumor cell dissemination in HER2+ BC.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Oncogénicas/metabolismo , Receptor ErbB-2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Terapia Molecular Dirigida , Proteínas Oncogénicas/genética , ARN Interferente Pequeño/genética , Receptor ErbB-2/genética , Receptores Acoplados a Proteínas G/genética , Reproducibilidad de los Resultados , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Breast Cancer Res Treat ; 157(2): 203-210, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27178335

RESUMEN

Atrophic vaginitis represents a major barrier to compliance with aromatase inhibitor (AI) therapy in breast cancer (BC) survivors. While local estrogen therapy is effective for postmenopausal vaginal dryness, the efficacy of such therapies has not been evaluated systematically in hormone receptor-positive (HR+) BC patients on AI therapy. Furthermore, the potential risk of breast cancer recurrence with vaginal estrogen therapy represents a long-term safety concern for the patients with HR + BC. Unfortunately, there is no standardized assay to measure very low concentrations of estradiol (E2) in these women being treated with AI therapy. This makes it difficult to evaluate even indirectly the potential risk of BC recurrence with vaginal estrogen therapy in HR + BC patients on AI therapy. In this review, we describe available assays to measure very low concentrations of E2, discuss the Food and Drug Administration-approved vaginal estrogen products on the market, and summarize published and ongoing clinical trials evaluating the safety and efficacy of vaginal estrogen in HR + BC patients on AI therapy. In the absence of any randomized controlled clinical trials, this review serves as a summary of available clinical data and ongoing studies to aid clinicians in selecting the best available option for their patients.


Asunto(s)
Inhibidores de la Aromatasa/efectos adversos , Vaginitis Atrófica/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Estrógenos/administración & dosificación , Recurrencia Local de Neoplasia/inducido químicamente , Administración Intravaginal , Inhibidores de la Aromatasa/uso terapéutico , Vaginitis Atrófica/sangre , Vaginitis Atrófica/inducido químicamente , Neoplasias de la Mama/sangre , Ensayos Clínicos como Asunto , Estradiol/sangre , Estrógenos/efectos adversos , Femenino , Humanos , Cooperación del Paciente , Posmenopausia , Sobrevivientes
12.
Breast Cancer Res ; 17: 3, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25572662

RESUMEN

INTRODUCTION: Real-time monitoring of biologic changes in tumors may be possible by investigating the transitional cells such as circulating tumor cells (CTCs) and disseminated tumor cells in bone marrow (BM-DTCs). However, the small numbers of CTCs and the limited access to bone marrow aspirates in cancer patients pose major hurdles. The goal of this study was to determine whether breast cancer (BC) patient-derived xenograft (PDX) mice could provide a constant and renewable source of CTCs and BM-DTCs, thereby representing a unique system for the study of metastatic processes. METHODS: CTCs and BM-DTCs, isolated from BC PDX-bearing mice, were identified by immunostaining for human pan-cytokeratin and nuclear counterstaining of red blood cell-lysed blood and bone marrow fractions, respectively. The rate of lung metastases (LM) was previously reported in these lines. Associations between the presence of CTCs, BM-DTCs, and LM were assessed by the Fisher's Exact and Cochran-Mantel-Haenszel tests. Two separate genetic signatures associated with the presence of CTC clusters and with lung metastatic potential were computed by using the expression arrays of primary tumors from different PDX lines and subsequently overlapped to identify common genes. RESULTS: In total, 18 BC PDX lines were evaluated. CTCs and BM-DTCs, present as either single cells or clusters, were detected in 83% (15 of 18) and 62.5% (10 to16) of the lines, respectively. A positive association was noted between the presence of CTCs and BM-DTCs within the same mice. LM was previously found in 9 of 18 (50%) lines, of which all nine had detectable CTCs. The presence of LM was strongly associated with the detection of CTC clusters but not with individual cells or detection of BM-DTCs. Overlapping of the two genetic signatures of the primary PDX tumors associated with the presence of CTC clusters and with lung metastatic potential identified four genes (HLA-DP1A, GJA1, PEG3, and XIST). This four-gene profile predicted distant metastases-free survival in publicly available datasets of early BC patients. CONCLUSION: This study suggests that CTCs and BM-DTCs detected in BC PDX-bearing mice may represent a valuable and unique preclinical model for investigating the role of these rare cells in tumor metastases.


Asunto(s)
Neoplasias de la Mama/patología , Células Neoplásicas Circulantes/patología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Neoplasias Pulmonares/secundario , Ratones , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/metabolismo , Pronóstico , Transcriptoma
13.
Breast Cancer Res Treat ; 153(3): 591-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26337685

RESUMEN

The febrile neutropenia (FN) rates reported with the docetaxel 75 mg/m(2) plus cyclophosphamide 600 mg/m(2) (TC) regimen given every 3 weeks vary from 4 to 69 % in early-stage breast cancer (ESBC) patients. This creates uncertainty as to whether patients receiving the TC regimen should also receive granulocyte colony-stimulating factor primary prophylaxis (G-CSFpp), which is recommended when chemotherapy regimens have ≥20 % FN rate. We conducted a meta-analysis of published studies to determine FN rate with the TC regimen, its dependence on patients' age, and the efficacy of G-CSFpp in reducing it in ESBC patients. We systematically searched the literature via PUBMED using the following terms: 'docetaxel', 'cyclophosphamide', 'febrile neutropenia', and 'breast cancer'. Inclusion criteria were full text peer-reviewed clinical studies in English reporting FN rates with TC regimen in relationship to G-CSFpp. Comprehensive meta-analysis software was used for all statistical analyses. Eight studies (N = 1542 patients) were included in our meta-analysis. The pooled mean FN rate was 23.2 % (95 % confidence interval (CI) 6.9-55.2 %; Q = 218.17, I (2) = 97.7). The FN risk in <65 years old patients was lower by 67.7 % compared to that in patients ≥65 years old (pooled odds ratio (OR) 0.323; 95 % CI 0.127-0.820; P = 0.017). The FN risk was reduced by 92.3 % with G-CSFpp (pooled OR 0.077; 95 % CI 0.013-0.460; P = 0.005). Our meta-analysis demonstrated that TC regimen was associated with ≥20 % FN risk, which was significantly higher in patients ≥65 years old and improved with G-CSFpp. G-CSFpp should be considered for all ESBC patients receiving TC regimen, especially those ≥65 years old.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/complicaciones , Neutropenia Febril/etiología , Neutropenia Febril/prevención & control , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/patología , Ciclofosfamida/administración & dosificación , Docetaxel , Neutropenia Febril/epidemiología , Femenino , Humanos , Estadificación de Neoplasias , Oportunidad Relativa , Profilaxis Pre-Exposición , Riesgo , Taxoides/administración & dosificación
14.
Acta Haematol ; 131(3): 133-40, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24192756

RESUMEN

Tumor cell contamination (TCC) of peripheral blood stem cells (PBSCs) is a major risk in the autologous PBSC transplant setting. However, the effect of different mobilization regimens (cytokines only versus cytokines + chemotherapy) on TCC of PBSCs and its impact on treatment outcomes have not been systematically reviewed. In the present meta-analysis, we aimed to investigate this effect in breast cancer patients since multiple studies have been conducted in this setting. We systematically searched MEDLINE and Cochrane Library up to May 2012. Seventeen studies (1,819 patients) were assessed. There was no significant difference in the incidence of TCC of PBSCs between the two mobilization regimens. When the analysis was restricted to granulocyte colony-stimulating factor as a cytokine, this difference was again not significant. We also found that TCC of PBSCs was associated with a higher annual recurrence rate in these patients. This suggests that there may be a significant risk for reinfusion of tumor cell-positive PBSCs, and whether it can increase the risk of disease recurrence needs to be determined. This study also raises important questions regarding the causes of TCC of PBSCs. These issues should be investigated systematically in PBSC transplant patients.


Asunto(s)
Neoplasias de la Mama/terapia , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre de Sangre Periférica/métodos , Antineoplásicos/administración & dosificación , Eliminación de Componentes Sanguíneos/efectos adversos , Eliminación de Componentes Sanguíneos/métodos , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Citocinas/administración & dosificación , Femenino , Movilización de Célula Madre Hematopoyética/efectos adversos , Humanos , Células Neoplásicas Circulantes/patología , Trasplante de Células Madre de Sangre Periférica/efectos adversos , Recurrencia , Factores de Riesgo , Trasplante Autólogo , Resultado del Tratamiento
15.
Cardiooncology ; 10(1): 17, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38532523

RESUMEN

BACKGROUND: Cardiovascular (CV) disease is a leading cause of death in breast cancer (BC) patients due to the increased age and treatments. While individual ß-blockers have been investigated to manage CV complications, various ß-blockers have not been compared for their effects on CV death in this population. We aimed to compare CV mortality in older BC patients taking one of the commonly used ß-blockers. METHODS: This retrospective cohort study was conducted using the Surveillance, Epidemiology and End Results (SEER) - Medicare data (2010-2015). Patients of age 66 years or older at BC diagnosis receiving metoprolol, atenolol, or carvedilol monotherapy were included. The competing risk regression model was used to determine the risk of CV mortality in the three ß-blocker groups. The multivariable model was adjusted for demographic and clinical covariates. The adjusted hazard ratio (HR) and 95% confidence intervals (CI) were reported for the risk of CV mortality. RESULTS: The study cohort included 6,540 patients of which 55% were metoprolol users, 30% were atenolol users, and 15% were carvedilol users. Metoprolol was associated with a 37% reduced risk of CV mortality (P = 0.03) compared to carvedilol after adjusting for the covariates (HR = 0.63; 95% CI 0.41-0.96). No significant difference in the risk of CV mortality between atenolol and carvedilol users was observed (HR = 0.74; 95% CI 0.44-1.22). CONCLUSIONS: Our findings suggest that metoprolol is associated with a reduced risk of CV mortality in BC patients. Future studies are needed to confirm these findings and understand the mechanism of action.

16.
Pharmacoeconomics ; 41(6): 709-718, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36920662

RESUMEN

BACKGROUND AND OBJECTIVE: Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors improve progression-free survival when combined with endocrine therapies in patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative metastatic breast cancer. However, the comparative cost effectiveness of utilizing three US Food and Drug Administration-approved CDK4/6 inhibitors is unknown. Therefore, we aimed to evaluate the cost effectiveness of individual CDK4/6 inhibitors (palbociclib, ribociclib, abemaciclib) with letrozole versus letrozole monotherapy in the first-line treatment of hormone receptor-positive/human epidermal growth factor receptor 2-negative metastatic breast cancer in the USA. METHODS: We constructed a Markov-based decision-analytic model to evaluate the cost effectiveness of CDK4/6 inhibitors plus endocrine therapies over a 40-year lifetime from a third-party payer perspective. The model incorporated health states (progression-free disease, progressive disease, and death), major adverse events (neutropenia), and cancer-specific and all-cause mortality. Using clinical efficacy and quality-of-life scores (utility) data from clinical trials, we estimated quality-adjusted life-years (QALYs) and incremental cost-effectiveness ratios using Medicare charges reported in US dollars per 2022 valuation and a discount rate of 3% applied to costs and outcomes. We performed deterministic and probabilistic sensitivity analyses to evaluate parametric and decision uncertainty. RESULTS: Compared to letrozole, the model estimated an increase of 5.72, 5.87, and 6.39 in QALYs and costs of $799,178, $788,168, and $741,102 in combining palbociclib, ribociclib, and abemaciclib plus letrozole, respectively. Palbociclib or ribociclib plus letrozole were dominated by abemaciclib plus letrozole. Compared with letrozole, abemaciclib plus letrozole resulted in an incremental cost-effectiveness ratio of $457,538 per QALY with an incremental cost of $553,621 and an incremental QALY gain of 1.21. The results were sensitive to the cost of abemaciclib, disease progression utility, and patients' age. CONCLUSIONS: At a willingness to pay of $100,000/QALY gained, our model predicts that combining CDK4/6 inhibitors plus letrozole is not cost effective with a marginal increase in QALYs at a high cost. Lowering the cost of these drugs or identifying patients who can receive maximal benefit from CDK4/6 inhibitors would improve the value of this regimen in patients.


Asunto(s)
Neoplasias de la Mama , Anciano , Humanos , Femenino , Estados Unidos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Letrozol/uso terapéutico , Análisis de Costo-Efectividad , Posmenopausia , Medicare , Protocolos de Quimioterapia Combinada Antineoplásica , Receptor ErbB-2/metabolismo , Quinasa 4 Dependiente de la Ciclina/uso terapéutico
17.
Int J Clin Pharm ; 45(6): 1396-1404, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37380914

RESUMEN

BACKGROUND: Adherence to oral endocrine therapy (OET) is crucial in ensuring its maximum benefit in the prevention and treatment of hormone receptor-positive (HR +) breast cancer (BC). Medication use behavior is suboptimal especially in racial/ethnic minorities with lower socioeconomic status (SES). AIM: We aimed to assess the impact of the coronavirus disease 2019 (COVID-19) pandemic on OET adherence and identify demographic and/or clinical characteristics associated with nonadherence in racial/ethnic minorities with lower SES. METHOD: A retrospective study was conducted at the Harris Health System in Houston, Texas. Data were collected during the 6 months before and 6 months after the start of the pandemic. The adherence was assessed using the prescription refill data using the proportion of days covered. A multivariable logistic regression model was used to identify demographic/clinical characteristics associated with nonadherence. Eighteen years or older patients on appropriate doses of OET for prevention or treatment of BC were included. RESULTS: In 258 patients, adherence was significantly lower during the pandemic (44%) compared to before the pandemic (57%). The demographic/clinical characteristics associated with OET nonadherence before the pandemic were Black/African American, obesity/extreme obesity, prevention setting, tamoxifen therapy, and 4 or more years on OET. During the pandemic, prevention setting and those not using home delivery were more likely to be nonadherent. CONCLUSION: OET adherence was significantly reduced during the COVID-19 pandemic in racial/ethnic minority patients with low SES. Patient-centered interventions are necessary to improve OET adherence in these patients.


Asunto(s)
Neoplasias de la Mama , COVID-19 , Humanos , Femenino , Minorías Étnicas y Raciales , Pandemias , Estudios Retrospectivos , Etnicidad , Estatus Socioeconómico Bajo , Cumplimiento de la Medicación , COVID-19/epidemiología , Grupos Minoritarios , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/epidemiología , Obesidad
18.
Cell Rep ; 42(8): 112821, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37467106

RESUMEN

Aberrant activation of the forkhead protein FOXA1 is observed in advanced hormone-related cancers. However, the key mediators of high FOXA1 signaling remain elusive. We demonstrate that ectopic high FOXA1 (H-FOXA1) expression promotes estrogen receptor-positive (ER+) breast cancer (BC) metastasis in a xenograft mouse model. Mechanistically, H-FOXA1 reprograms ER-chromatin binding to elicit a core gene signature (CGS) enriched in ER+ endocrine-resistant (EndoR) cells. We identify Secretome14, a CGS subset encoding ER-dependent cancer secretory proteins, as a strong predictor for poor outcomes of ER+ BC. It is elevated in ER+ metastases vs. primary tumors, irrespective of ESR1 mutations. Genomic ER binding near Secretome14 genes is also increased in mutant ER-expressing or mitogen-treated ER+ BC cells and in ER+ metastatic vs. primary tumors, suggesting a convergent pathway including high growth factor receptor signaling in activating pro-metastatic secretome genes. Our findings uncover H-FOXA1-induced ER reprogramming that drives EndoR and metastasis partly via an H-FOXA1/ER-dependent secretome.

19.
Breast Cancer Res ; 14(3): 310, 2012 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-22621279

RESUMEN

One of the possible mechanisms contributing to the intrinsic resistance of cancer stem cells (CSCs) to conventional therapies is the inefficiency of activating the apoptotic machinery. In a recent study by Lang and colleagues, the engineered constitutively active pro-apoptotic protein BikDD, which works by inhibiting multiple Bcl-2 family members, was tested in various preclinical breast cancer models. Delivered to cells via an innovative cancer cell-specific gene-therapy approach, BikDD showed potent activity against CSCs and synergized with lapatinib and paclitaxel treatment. This novel and promising therapy warrants further translation to the clinic.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de la Membrana/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/uso terapéutico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Antígeno CD24/efectos de los fármacos , Femenino , Humanos , Receptores de Hialuranos/efectos de los fármacos , Lapatinib , Proteínas de la Membrana/uso terapéutico , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Mitocondriales , Células Madre Neoplásicas/metabolismo , Paclitaxel/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/genética , Quinazolinas/uso terapéutico , Interferencia de ARN , ARN Interferente Pequeño
20.
Ann Pharmacother ; 46(6): 802-11, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22619477

RESUMEN

OBJECTIVE: To review the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of eribulin (Halaven). DATA SOURCES: A literature search (up to December 2011) using the terms eribulin, Halaven, ER-086526, and E7389 was performed with PubMed, Google Scholar, selected Ovid bibliography searches, and the abstract search tool from the American Society of Clinical Oncology Annual Meetings and the San Antonio Breast Cancer Symposia. Additional references from the bibliographies of these articles were also assessed. DATA EXTRACTION: English-language preclinical and clinical studies on the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of eribulin were reviewed. DATA SYNTHESIS: Eribulin is a novel microtubule inhibitor with a unique mechanism of action, which involves interaction with a distinct binding site on ß-tubulin leading to G(2)/M phase cell-cycle arrest and apoptosis. Eribulin has been approved by the Food and Drug Administration for the treatment of metastatic breast cancer in patients who have been previously treated with an anthracycline and a taxane. In a pivotal Phase 3 study conducted in patients with metastatic breast cancer, eribulin 1.4 mg/m(2), administered over 2-5 minutes as an intravenous infusion on days 1 and 8 of 21-day cycles, was associated with a significantly increased median overall survival of 13.1 months compared to the median overall survival of 10.6 months in the therapy of physician's choice. Eribulin has also shown activity in Phase 2 studies in other types of cancers, such as non-small cell lung cancer, prostate cancer, urothelial cancer, soft tissue sarcomas, and platinum-susceptible ovarian, fallopian tube, or peritoneal cancers. The most severe (grade 3/4) adverse effects associated with eribulin include neutropenia, leukopenia, and peripheral neuropathy. Common toxicities include fatigue, neutropenia, alopecia, anemia, and peripheral neuropathy. CONCLUSIONS: Eribulin is a promising new cytotoxic chemotherapy agent due to its ability to treat cancers that are refractory or resistant to other drugs as well as its manageable toxicity profile.


Asunto(s)
Antineoplásicos/uso terapéutico , Citotoxinas/uso terapéutico , Furanos/uso terapéutico , Cetonas/uso terapéutico , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacocinética , Citotoxinas/farmacocinética , Furanos/farmacocinética , Humanos , Cetonas/farmacocinética , Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA