Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Theranostics ; 12(2): 639-656, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34976205

RESUMEN

Rationale: B cells have emerged as key regulators in protective cancer immunity. However, the activation pathways induced in B cells during effective immunotherapy are not well understood. Methods: We used a novel localized ablative immunotherapy (LAIT), combining photothermal therapy (PTT) with intra-tumor delivery of the immunostimulant N-dihydrogalactochitosan (GC), to treat mice bearing mouse mammary tumor virus-polyoma middle tumor-antigen (MMTV-PyMT). We used single-cell RNA sequencing to compare the transcriptional changes induced by PTT, GC and PTT+GC in B cells within the tumor microenvironment (TME). Results: LAIT significantly increased survival in the tumor-bearing mice, compared to the treatment by PTT and GC alone. We found that PTT, GC and PTT+GC increased the proportion of tumor-infiltrating B cells and induced gene expression signatures associated with B cell activation. Both GC and PTT+GC elevated gene expression associated with antigen presentation, whereas GC elevated transcripts that regulate B cell activation and GTPase function and PTT+GC induced interferon response genes. Trajectory analysis, where B cells were organized according to pseudotime progression, revealed that both GC and PTT+GC induced the differentiation of B cells from a resting state towards an effector phenotype. The analyses confirmed upregulated interferon signatures in the differentiated tumor-infiltrating B cells following treatment by PTT+GC but not by GC. We also observed that breast cancer patients had significantly longer survival time if they had elevated expression of genes in B cells that were induced by PTT+GC therapy in the mouse tumors. Conclusion: Our findings show that the combination of local ablation and local application of immunostimulant initiates the activation of interferon signatures and antigen-presentation in B cells which is associated with positive clinical outcomes for breast cancer. These findings broaden our understanding of LAIT's regulatory roles in remodeling TME and shed light on the potentials of B cell activation in clinical applications.


Asunto(s)
Presentación de Antígeno , Linfocitos B/inmunología , Inmunoterapia , Interferones/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Animales , Linfocitos B/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/terapia , Ratones , Transcriptoma
2.
Cancer Prev Res (Phila) ; 12(8): 493-506, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31266827

RESUMEN

Physical activity and the prevention of weight gain decrease breast cancer incidence and improve survival. Unraveling the biological mechanisms underlying these cancer prevention effects is difficult because activity and dietary restriction are often linked. The goal of this study was to determine whether physical activity (PA), preventing weight gain via energy restriction (ER), or the combination was most effective in delaying tumor growth, reducing metastatic progression, and improving survival in the 4T1.2 mammary tumor model. Furthermore, we determined whether any of these interventions prevented the expansion of protumor immunosuppressive cells and altered the tumor microenvironment (TME). Female BALB/c mice (n = 7-20/group) were randomized to sedentary (SED) or PA wheel cages and fed ad libitum (AL) or 90% of control food intake (ER). After 8 weeks on the interventions, mice were inoculated with 5 × 104 4T1.2luc cells into the 4th mammary fat pad and continued on their respective intervention. PA+ER significantly delayed primary tumor growth (final tumor volume, 0.193 ± 0.042 vs. 0.369 ± 0.049 cm3, P < 0.001), reduced metastatic burden in the lungs (0.72 ± 0.36 vs. 16.27 ± 6.98, P = 0.054) and increased survival (median survival, 68 vs 40 days, P = 0.043) compared with SED+AL mice. PA+ER also reduced the expression level of metastatic and immunosuppressive genes and resulted in favorable changes in immune cell infiltrates in the tumor. These data suggest that both PA and ER are needed to reduce tumor growth, delay metastatic progression, and improve survival, and that this protection is associated with changes in immune-mediated mechanisms.


Asunto(s)
Restricción Calórica , Neoplasias Mamarias Experimentales/terapia , Condicionamiento Físico Animal/fisiología , Conducta Sedentaria , Microambiente Tumoral/inmunología , Animales , Conducta Animal , Línea Celular Tumoral/trasplante , Progresión de la Enfermedad , Femenino , Humanos , Glándulas Mamarias Animales/inmunología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones , Células Supresoras de Origen Mieloide/inmunología
3.
BMC Cancer ; 19(1): 536, 2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164094

RESUMEN

BACKGROUND: Aerobic exercise has been shown to slow tumor progression in rodents and humans, but the mechanisms behind this effect are still unclear. Here we show that aerobic exercise in the form of chronic endurance training suppresses tumor recruitment of FoxP3+ Treg cells thus enhancing antitumor immune efficiency. METHODS: Adult wild-type and athymic BALB/c female mice were endurance-trained for 8 weeks. Circulating leukocytes as well as muscle and liver mtDNA copy number were compared to aged-matched concurrent sedentary controls to establish systemic effects. 4 T1 murine mammary tumor cells were injected subcutaneously to the 4th mammary pad at the end of the training period. Tumor growth and survival rates were compared, together with antitumor immune response. RESULTS: Exercised wild-type had 17% slower growth rate, 24% longer survival, and 2-fold tumor-CD+ 8/FoxP3+ ratio than sedentary controls. Exercised athymic BALB/c females showed no difference in tumor growth or survival rates when compared to sedentary controls. CONCLUSIONS: Cytotoxic T cells are a significant factor in endurance exercise-induced suppression of tumor growth. Endurance exercise enhances antitumor immune efficacy by increasing intratumoral CD8+/FoxP3+ ratio.


Asunto(s)
Progresión de la Enfermedad , Entrenamiento Aeróbico , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/fisiopatología , Condicionamiento Físico Animal , Linfocitos T Reguladores/metabolismo , Animales , Línea Celular Tumoral , Femenino , Factores de Transcripción Forkhead/metabolismo , Estimación de Kaplan-Meier , Ácido Láctico/sangre , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proyectos Piloto , Tasa de Supervivencia , Linfocitos T Citotóxicos/metabolismo , Carga Tumoral
4.
Breast Cancer Res ; 21(1): 41, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30867005

RESUMEN

BACKGROUND: Obesity is associated with an increased risk of breast cancer recurrence and cancer death. Recurrent cancers arise from the pool of residual tumor cells, or minimal residual disease (MRD), that survives primary treatment and persists in the host. Whether the association of obesity with recurrence risk is causal is unknown, and the impact of obesity on MRD and breast cancer recurrence has not been reported in humans or in animal models. METHODS: Doxycycline-inducible primary mammary tumors were generated in intact MMTV-rtTA;TetO-HER2/neu (MTB/TAN) mice or orthotopic recipients fed a high-fat diet (HFD; 60% kcal from fat) or a control low-fat diet (LFD; 10% kcal from fat). Following oncogene downregulation and tumor regression, mice were followed for clinical recurrence. Body weight was measured twice weekly and used to segregate HFD mice into obese (i.e., responders) and lean (i.e., nonresponders) study arms, and obesity was correlated with body fat percentage, glucose tolerance (measured using intraperitoneal glucose tolerance tests), serum biomarkers (measured by enzyme-linked immunosorbent assay), and tissue transcriptomics (assessed by RNA sequencing). MRD was quantified by droplet digital PCR. RESULTS: HFD-Obese mice weighed significantly more than HFD-Lean and LFD control mice (p < 0.001) and had increased body fat percentage (p < 0.001). Obese mice exhibited fasting hyperglycemia, hyperinsulinemia, and impaired glucose tolerance, as well as decreased serum levels of adiponectin and increased levels of leptin, resistin, and insulin-like growth factor 1. Tumor recurrence was accelerated in HFD-Obese mice compared with HFD-Lean and LFD control mice (median relapse-free survival 53.0 days vs. 87.0 days vs. 80.0 days, log-rank p < 0.001; HFD-Obese compared with HFD-Lean HR 2.52, 95% CI 1.52-4.16; HFD-Obese compared with LFD HR 2.27, 95% CI 1.42-3.63). HFD-Obese mice harbored a significantly greater number of residual tumor cells than HFD-Lean and LFD mice (12,550 ± 991 vs. 7339 ± 2182 vs. 4793 ± 1618 cells, p < 0.001). CONCLUSION: These studies provide a genetically engineered mouse model for study of the association of diet-induced obesity with breast cancer recurrence. They demonstrate that this model recapitulates physiological changes characteristic of obese patients, establish that the association between obesity and recurrence risk is causal in nature, and suggest that obesity is associated with the increased survival and persistence of residual tumor cells.


Asunto(s)
Neoplasias de la Mama/mortalidad , Neoplasias Mamarias Experimentales/patología , Recurrencia Local de Neoplasia/patología , Obesidad/patología , Animales , Índice de Masa Corporal , Peso Corporal , Neoplasias de la Mama/patología , Línea Celular Tumoral/trasplante , Conjuntos de Datos como Asunto , Dieta Alta en Grasa/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/mortalidad , Ratones Obesos , Ratones Transgénicos , Recurrencia Local de Neoplasia/mortalidad , Neoplasia Residual , Obesidad/etiología , Receptor ErbB-2/genética , Análisis de Supervivencia
5.
Cancer Immunol Res ; 6(12): 1561-1577, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30341213

RESUMEN

Immune-checkpoint inhibition (ICI) has revolutionized treatment in cancers that are naturally immunogenic by enabling infiltration of T cells into the tumor microenvironment (TME) and promoting cytotoxic signaling pathways. Tumors possessing complex immunosuppressive TMEs such as breast and pancreatic cancers present unique therapeutic obstacles as response rates to ICI remain low. Such tumors often recruit myeloid-derived suppressor cells (MDSCs), whose functioning prohibits both T-cell activation and infiltration. We attempted to sensitize these tumors to ICI using epigenetic modulation to target MDSC trafficking and function to foster a less immunosuppressive TME. We showed that combining a histone deacetylase inhibitor, entinostat (ENT), with anti-PD-1, anti-CTLA-4, or both significantly improved tumor-free survival in both the HER2/neu transgenic breast cancer and the Panc02 metastatic pancreatic cancer mouse models. Using flow cytometry, gene-expression profiling, and ex vivo functional assays, we characterized populations of tumor-infiltrating lymphocytes (TILs) and MDSCs, as well as their functional capabilities. We showed that addition of ENT to checkpoint inhibition led to significantly decreased suppression by granulocytic MDSCs in the TME of both tumor types. We also demonstrated an increase in activated granzyme-B-producing CD8+ T effector cells in mice treated with combination therapy. Gene-expression profiling of both MDSCs and TILs identified significant changes in immune-related pathways. In summary, addition of ENT to ICI significantly altered infiltration and function of innate immune cells, allowing for a more robust adaptive immune response. These findings provide a rationale for combination therapy in patients with immune-resistant tumors, including breast and pancreatic cancers.


Asunto(s)
Benzamidas/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Células Supresoras de Origen Mieloide/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Piridinas/farmacología , Animales , Antineoplásicos/farmacología , Antígeno CTLA-4/antagonistas & inhibidores , Carcinoma Ductal Pancreático/mortalidad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Masculino , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Supresoras de Origen Mieloide/inmunología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
6.
Int J Nanomedicine ; 12: 8337-8351, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29200846

RESUMEN

An intraoperative technique to accurately identify microscopic tumor residuals could decrease the risk of positive surgical margins. Several lines of evidence support the expression and immunotherapeutic effect of PD-1 in breast cancer. Here, we sought to develop a fluorescence-labeled PD-1 probe for in vivo breast tumor imaging and image-guided surgery. The efficacy of PD-1 monoclonal antibody (PD-1 mAb) as adjuvant immunotherapy after surgery was also assessed. PD-1-IRDye800CW was developed and examined for its application in tumor imaging and image-guided tumor resection in an immunocompetent 4T1 mouse tumor model. Fluorescence molecular imaging was performed to monitor probe biodistribution and intraoperative imaging. Bioluminescence imaging was performed to monitor tumor growth and evaluate postsurgical tumor residuals, recurrences, and metastases. The PD-1-IRDye800CW exhibited a specific signal at the tumor region compared with the IgG control. Furthermore, PD-1-IRDye800CW-guided surgery combined with PD-1 adjuvant immunotherapy inhibited tumor regrowth and microtumor metastases and thus improved survival rate. Our study demonstrates the feasibility of using PD-1-IRDye800CW for breast tumor imaging and image-guided tumor resection. Moreover, PD-1 mAb adjuvant immunotherapy reduces cancer recurrences and metastases emanating from tumor residuals.


Asunto(s)
Colorantes Fluorescentes/química , Neoplasias Mamarias Experimentales/terapia , Monitoreo Intraoperatorio/métodos , Receptor de Muerte Celular Programada 1/química , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/metabolismo , Femenino , Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/uso terapéutico , Inmunoterapia/métodos , Mediciones Luminiscentes/métodos , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/cirugía , Ratones Endogámicos BALB C , Imagen Molecular/métodos , Monitoreo Intraoperatorio/instrumentación , Imagen Óptica/métodos , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Cirugía Asistida por Computador/métodos , Distribución Tisular
7.
Nutr Cancer ; 69(7): 1036-1042, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28937793

RESUMEN

BACKGROUND: One major concern in the treatment of cancer patients during chemotherapy is drug resistance. Here we investigated the effects of soy isoflavone extracts alone or in combination with Docetaxel on the drug resistance, angiogenesis, apoptosis, and tumor volume in mouse 4T1 breast tumor model. METHODS: Sixty female BALB/c mice were randomly divided into 4 groups: control, dietary soy isoflavone extract [Iso, 100 mg/kg diet (0.01%)], Docetaxel (10 mg/kg) injection, and the combination of dietary soy isoflavone extract and intravenous Docetaxel injection (Docetaxel + Iso). One week after the third injection, the breast tumors of eight mice from each group were excised to analyze NF-κBp65' vascular endothelial growth factor receptor-2 (VEGFR2) and Pgp gene and protein expressions and the other seven mice were monitored for survival rate analysis until they died. RESULTS: NF-κBp65 gene and protein expressions were significantly lower in the Docetaxel + Iso group in comparison with that of the Docetaxel group. VEGFR2 protein expression in the Docetaxel + Iso and Iso groups was significantly lower than that of the Docetaxel group. CONCLUSION: These findings may indicate that the combined use of isoflavone extracts together with chemotherapeutic agents has more efficient anti-carcinogenic effects than their individual use.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Glycine max/química , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Extractos Vegetales/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Docetaxel , Ingestión de Alimentos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Isoflavonas/química , Isoflavonas/farmacología , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Tasa de Supervivencia , Taxoides/administración & dosificación , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Cancer Lett ; 389: 49-58, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28041977

RESUMEN

The induction of localized pro-inflammatory niches in the periphery is instrumental in metastasis. In order to better understand how tumors engage distal sites and activate a pro-inflammatory response we utilized syngeneic breast cancers as a model and showed that soluble factors from the neoplastic epithelium activate the expression of the monocyte chemoattractive protein (MCP) chemokines of the mouse 11C cluster that include Ccl1, Ccl2, Ccl7, Ccl8, Ccl11 and Ccl12. Tissues such as the lungs and the brain, that are more prone to colonization by breast cancer cells, were more sensitive to MCP cluster chemokine induction than others such as the liver. Subsequent analyses involving chemokine arrays in breast cancer cells and media followed by functional validation assays in in vitro and in vivo identified the cytokine Ccl3 as the principle mediator of the communication between the neoplastic epithelium and the peripheral tissues in terms of MCP cluster chemokine induction. Our results show that MCP chemokines are activated in peripheral tissues of breast cancer-bearing mice, by a mechanism that involves breast cancer cell-derived Ccl3. Interference with the expression of cancer cell-derived Ccl3 may find application in the management of breast cancer metastases.


Asunto(s)
Quimiocina CCL3/fisiología , Neoplasias Mamarias Experimentales/inmunología , Proteínas Quimioatrayentes de Monocitos/biosíntesis , Animales , Femenino , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Metástasis de la Neoplasia
9.
Oncol Rep ; 37(1): 368-378, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27878284

RESUMEN

Manumycin A is a natural antibiotic isolated from Streptomyces parvulus with broad range of biological activities including antineoplastic activity in several in vitro and in vivo cancer models. Immodin [dialyzable leukocyte extract (DLE)] is a dialysate released from disintegrated blood leukocytes of healthy donors which exerts immunonormalizing effects on cell-mediated immune responses. The aim of the present study was to explore the antitumor potential of the combination of manumycin A and Immodin in an experimental breast cancer model. Experiments were carried using a 4T1 tumor-bearing BALB/c mouse model. Survival analysis, tumor growth, hematological and biochemical profiles, leukocyte differential, phagocytic activity of leukocytes and histology of the primary tumor were examined. The combination treatment suppressed the tumor growth and prolonged the survival of tumor-bearing mice, decreased the number of monocytes, plateletes and plateletcrit in peripheral blood of the tumor-bearing mice and increased the infiltration of neutrophils and eosinophils in the primary tumor. Moreover, individual therapies enhanced the phagocytic activity of monocytes and neutrophils. These findings demonstrate the antitumor effect of the combination of manumycin A and Immodin in 4T1 tumor-bearing mice associated with strong antiplatelet activity and innate immunity activation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Leucocitos/química , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Animales , Femenino , Granulocitos/efectos de los fármacos , Granulocitos/patología , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/farmacología , Recuento de Leucocitos , Neoplasias Mamarias Experimentales/mortalidad , Ratones Endogámicos BALB C , Fagocitosis/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Polienos/administración & dosificación , Alcamidas Poliinsaturadas/administración & dosificación , Análisis de Supervivencia
10.
Cell Rep ; 16(8): 2087-2101, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27524621

RESUMEN

Invasive lobular carcinoma (ILC) is an aggressive breast cancer subtype with poor response to chemotherapy. Besides loss of E-cadherin, a hallmark of ILC, genetic inactivation of PTEN is frequently observed in patients. Through concomitant Cre-mediated inactivation of E-cadherin and PTEN in mammary epithelium, we generated a mouse model of classical ILC (CLC), the main histological ILC subtype. While loss of E-cadherin induced cell dissemination and apoptosis, additional PTEN inactivation promoted cell survival and rapid formation of invasive mammary tumors that recapitulate the histological and molecular features, estrogen receptor (ER) status, growth kinetics, metastatic behavior, and tumor microenvironment of human CLC. Combined inactivation of E-cadherin and PTEN is sufficient to cause CLC development. These CLCs showed significant tumor regression upon BEZ235-mediated inhibition of PI3K signaling. In summary, this mouse model provides important insights into CLC development and suggests inhibition of phosphatidylinositol 3-kinase (PI3K) signaling as a potential therapeutic strategy for targeting CLC.


Asunto(s)
Cadherinas/genética , Carcinoma Lobular/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Neoplasias Mamarias Experimentales/genética , Fosfohidrolasa PTEN/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Animales , Antineoplásicos/farmacología , Cadherinas/deficiencia , Carcinoma Lobular/tratamiento farmacológico , Carcinoma Lobular/mortalidad , Carcinoma Lobular/patología , Línea Celular Tumoral , Supervivencia Celular , Femenino , Perfilación de la Expresión Génica , Imidazoles/farmacología , Integrasas/genética , Integrasas/metabolismo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Invasividad Neoplásica , Fosfohidrolasa PTEN/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Quinolinas/farmacología , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Transducción de Señal , Análisis de Supervivencia , Microambiente Tumoral
11.
PLoS One ; 11(7): e0159686, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27433802

RESUMEN

MicroRNAs have emerged as ubiquitous post-transcriptional regulators that coordinate many fundamental processes within cells, including those commonly linked to cancer when dysregulated. Profiling microRNAs across stages of cancer progression provides focus as to which microRNAs are key players in cancer development and are therefore important to manipulate with interventions to delay cancer onset and progression. Calorie restriction is one of the most effective preventive interventions across many types of cancer, although its effects on microRNAs have not been well characterized. We used the dimethylbenz[a]-anthracene-induced model of luminal mammary cancer in Sprague Dawley rats to elucidate which microRNAs are linked to progression in this type of cancer and, subsequently, to study how calorie restriction affects such microRNAs. We identified eight microRNAs (miR-10a, miR-10b, miR-21, miR-124, miR-125b, miR-126, miR-145 and miR-200a) to be associated with DMBA-induced mammary tumor progression. Calorie restriction, which greatly increased tumor-free survival and decreased the overall size of tumors that did develop, significantly decreased the expression of one microRNA, miR-200a, which was positively associated with tumor progression. We further showed that inhibition of miR-200a function, mimicking the effect of calorie restriction on this microRNA, inhibited proliferation in both rat (LA7) and human (MCF7) luminal mammary cancer cell lines. These findings present, for the first time, a stage-specific profile of microRNAs in a rodent model of luminal mammary cancer. Furthermore, we have identified the regulation of miR-200a, a microRNA that is positively associated with progression in this model, as a possible mechanism contributing to the anticancer effects of calorie restriction.


Asunto(s)
Restricción Calórica , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , MicroARNs/genética , 9,10-Dimetil-1,2-benzantraceno , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Células MCF-7 , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/dietoterapia , Neoplasias Mamarias Experimentales/mortalidad , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Oligorribonucleótidos Antisentido/genética , Oligorribonucleótidos Antisentido/metabolismo , Ratas , Ratas Sprague-Dawley , Carga Tumoral
12.
Nat Commun ; 7: 11838, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-27319780

RESUMEN

The application of nanoparticle drug formulations, such as nanoliposomal doxorubicin (Doxil), is increasingly integrated in clinical cancer care. Despite nanomedicine's remarkable potential and growth over the last three decades, its clinical benefits for cancer patients vary. Here we report a non-invasive quantitative positron emission tomography (PET) nanoreporter technology that is predictive of therapeutic outcome in individual subjects. In a breast cancer mouse model, we demonstrate that co-injecting Doxil and a Zirconium-89 nanoreporter ((89)Zr-NRep) allows precise doxorubicin (DOX) quantification. Importantly, (89)Zr-NRep uptake also correlates with other types of nanoparticles' tumour accumulation. (89)Zr-NRep PET imaging reveals remarkable accumulation heterogeneity independent of tumour size. We subsequently demonstrate that mice with >25 mg kg(-1) DOX accumulation in tumours had significantly better growth inhibition and enhanced survival. This non-invasive imaging tool may be developed into a robust inclusion criterion for patients amenable to nanotherapy.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/análogos & derivados , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Nanomedicina Teranóstica/métodos , Circonio/administración & dosificación , Animales , Antibióticos Antineoplásicos/farmacocinética , Transporte Biológico , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Esquema de Medicación , Femenino , Humanos , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología , Radiofármacos/administración & dosificación , Análisis de Supervivencia , Distribución Tisular
13.
J Steroid Biochem Mol Biol ; 163: 129-35, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27142456

RESUMEN

SM6Met, a phytoestrogenic extract of Cyclopia subternata indigenous to the Western Cape province of South Africa, displays estrogenic attributes with potential for breast cancer chemoprevention. In this study, we report that SM6Met, in the presence of estradiol, induces a significant cell cycle G0/G1 phase arrest similar to the selective estrogen receptor modulator, tamoxifen. Furthermore, as a proof of concept, in the N-Methyl-N-nitrosourea induced rat mammary gland carcinogenesis model, SM6Met increases tumor latency by 7days and median tumor free survival by 42 days, while decreasing palpable tumor frequency by 32%, tumor mass by 40%, and tumor volume by 53%. Therefore, the current study provides proof of concept that SM6Met has definite potential as a chemopreventative agent against the development and progression of breast cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Cyclopia (Planta)/química , Glándulas Mamarias Animales/efectos de los fármacos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Fitoestrógenos/farmacología , Animales , Antineoplásicos Fitogénicos/aislamiento & purificación , Línea Celular Tumoral , Antagonistas de Estrógenos/farmacología , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Humanos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Metilnitrosourea , Fitoestrógenos/aislamiento & purificación , Ratas , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Análisis de Supervivencia , Tamoxifeno/farmacología , Carga Tumoral/efectos de los fármacos
14.
J Clin Invest ; 126(1): 99-111, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26595815

RESUMEN

Magnetic resonance-guided focused ultrasound (MRgFUS) facilitates noninvasive image-guided conformal thermal therapy of cancer. Yet in many scenarios, the sensitive tissues surrounding the tumor constrain the margins of ablation; therefore, augmentation of MRgFUS with chemotherapy may be required to destroy remaining tumor. Here, we used 64Cu-PET-CT, MRI, autoradiography, and fluorescence imaging to track the kinetics of long-circulating liposomes in immunocompetent mammary carcinoma-bearing FVB/n and BALB/c mice. We observed a 5-fold and 50-fold enhancement of liposome and drug concentration, respectively, within MRgFUS thermal ablation-treated tumors along with dense accumulation within the surrounding tissue rim. Ultrasound-enhanced drug accumulation was rapid and durable and greatly increased total tumor drug exposure over time. In addition, we found that the small molecule gadoteridol accumulates around and within ablated tissue. We further demonstrated that dilated vasculature, loss of vascular integrity resulting in extravasation of blood cells, stromal inflammation, and loss of cell-cell adhesion and tissue architecture all contribute to the enhanced accumulation of the liposomes and small molecule probe. The locally enhanced liposome accumulation was preserved even after a multiweek protocol of doxorubicin-loaded liposomes and partial ablation. Finally, by supplementing ablation with concurrent liposomal drug therapy, a complete and durable response was obtained using protocols for which a sub-mm rim of tumor remained after ablation.


Asunto(s)
Doxorrubicina/farmacocinética , Ultrasonido Enfocado de Alta Intensidad de Ablación , Neoplasias Mamarias Experimentales/terapia , Animales , Autorradiografía , Doxorrubicina/administración & dosificación , Femenino , Liposomas/farmacocinética , Imagen por Resonancia Magnética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/mortalidad , Ratones , Ratones Endogámicos BALB C , Tomografía de Emisión de Positrones , Distribución Tisular
15.
Mater Sci Eng C Mater Biol Appl ; 53: 68-75, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26042692

RESUMEN

BACKGROUND: Paclitaxel (PTX) is a first line chemotherapy drug for breast cancer. There have been few studies reported concerning the therapeutic efficacy of paclitaxel-conjugated polymeric micelles in breast cancer in vivo. METHODS: Two kinds of PTX conjugate micelles, one of which (M(PTX)) contained 25 wt.% of PTX and the other (M(FA/PTX)) contained 22.5 wt.% of PTX and 1.4 wt.% of folate (FA), were prepared for cell apoptosis and anti-tumor activity evaluation on EMT-6 mice breast cancer models in comparison with 0.9 wt.% saline (control) and equivalent PTX. Cell apoptosis was analyzed by flow cytometry. Breast tumors were examined histologically with H&E staining and immunohistochemically by examining Bax and Bcl-2 expression. The survival status of tumor-bearing mice with different treatments was also examined. RESULTS: On day 5 of the drug administration, the average tumor masses were 0.49, 0.33, 0.22, and 0.18 g for the control, PTX, M(PTX) and M(FA/PTX) groups, respectively. The inhibition rates of tumor growth calculated for the three drug groups were 32.6%, 51.6% and 62.3%, respectively. The percentage of cell apoptosis based on flow cytometry was 1.0%, 36.6%, 55.9% and 66.1%, respectively, which showed statistically significant differences (p<0.05) between three drug groups and the control group. Bcl-2 expression of PTX and M(FA/PTX) groups was lower than control group (p<0.05). Bax expression of drug groups was higher than control group (p<0.05). At an equivalent paclitaxel dose of 26.7 mg/kg, the average survival time was 33 days, 31 days, 34 days and 42 days, respectively (p<0.05). CONCLUSION: The M(FA/PTX) have better anti-tumor activity and are promising in treatment of human breast cancers.


Asunto(s)
Antineoplásicos , Sistemas de Liberación de Medicamentos/métodos , Ácido Fólico/química , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Paclitaxel , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Micelas , Paclitaxel/química , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Polietilenglicoles/química
16.
Eur J Gynaecol Oncol ; 36(2): 197-202, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26050360

RESUMEN

OBJECTIVE: This study aims to investigate the protective and sensitive effects of melatonin (MLT) in the treatment of breast cancer. MATERIALS AND METHODS: ER+ breast cancer rat model was established and then rats were randomly divided into five different groups as follows: control group, Diss group, adriamycin (ADM) group, MLT group, and MLT combined with adriamycin (M+A) group. Tumor weights and one month survival rate were compared among these groups. In addition, changes of tumor tissues and expression of E-cadherin were observed under optical microscopy or electro-microscopy. RESULTS: Tumor weights were significantly lighter in M+A group than those in ADM group (p < 0.05). Under optical and electro-microscopy, tumor cell apoptosis was obviously increased in MLT group, and tumor cell injury was more severe in M+A group than that in ADM group; additionally, expression of E-cadherin was higher in MLT group and M+A group than that in other groups. Moreover, MLT group had the highest one month survival rate (100%), there was the poorest life quality in ADM group, but the best life quality in MLT. CONCLUSION: MLT could enhance the sensitivity of tumor to ADM in vivo and improve patient's life quality.


Asunto(s)
Doxorrubicina/administración & dosificación , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Melatonina/administración & dosificación , Receptores de Estrógenos/análisis , Animales , Cadherinas/análisis , Femenino , Neoplasias Mamarias Experimentales/química , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratas , Ratas Sprague-Dawley
17.
Mol Cancer Ther ; 14(8): 1951-61, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25934710

RESUMEN

The anticancer actions of vitamin D and its hormonally active form, calcitriol, have been extensively documented in clinical and preclinical studies. However, the mechanisms underlying these actions have not been completely elucidated. Here, we examined the effect of dietary vitamin D and calcitriol on mouse breast tumor-initiating cells (TICs, also known as cancer stem cells). We focused on MMTV-Wnt1 mammary tumors, for which markers for isolating TICs have previously been validated. We confirmed that these tumors expressed functional vitamin D receptors and estrogen receptors (ER) and exhibited calcitriol-induced molecular responses including ER downregulation. Following orthotopic implantation of MMTV-Wnt1 mammary tumor cells into mice, calcitriol injections or a vitamin D-supplemented diet caused a striking delay in tumor appearance and growth, whereas a vitamin D-deficient diet accelerated tumor appearance and growth. Calcitriol inhibited TIC tumor spheroid formation in a dose-dependent manner in primary cultures and inhibited TIC self-renewal in secondary passages. A combination of calcitriol and ionizing radiation inhibited spheroid formation more than either treatment alone. Further, calcitriol significantly decreased TIC frequency as evaluated by in vivo limiting dilution analyses. Calcitriol inhibition of TIC spheroid formation could be overcome by the overexpression of ß-catenin, suggesting that the inhibition of Wnt/ß-catenin pathway is an important mechanism mediating the TIC inhibitory activity of calcitriol in this tumor model. Our findings indicate that vitamin D compounds target breast TICs reducing tumor-initiating activity. Our data also suggest that combining vitamin D compounds with standard therapies may enhance anticancer activity and improve therapeutic outcomes.


Asunto(s)
Calcitriol/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Vitamina D/farmacología , Animales , Peso Corporal , Calcio/sangre , Línea Celular Tumoral , Estrógenos/metabolismo , Femenino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones , Células Madre Neoplásicas/metabolismo , Receptores de Calcitriol/metabolismo , Receptores de Estrógenos/metabolismo , Carga Tumoral , Vitamina D/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
18.
Tumour Biol ; 36(5): 3325-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25528215

RESUMEN

Breast cancer is one of the most prevalent cancer types among women. The use of magnetic fluids for specific delivery of drugs represents an attractive platform for chemotherapy. In our previous studies, it was demonstrated that maghemite nanoparticles coated with rhodium (II) citrate (Magh-Rh2Cit) induced in vitro cytotoxicity and in vivo antitumor activity, followed by intratumoral administration in breast carcinoma cells. In this study, our aim was to follow intravenous treatment to evaluate the systemic antitumor activity and toxicity induced by these formulations in Balb/c mice bearing orthotopic 4T1 breast carcinoma. Female Balb/c mice were evaluated with regard to toxicity of intravenous treatments through analyses of hemogram, serum levels of alanine aminotransferase, iron, and creatinine and liver, kidney, and lung histology. The antitumor activity of rhodium (II) citrate (Rh2Cit), Magh-Rh2Cit, and maghemite nanoparticles coated with citrate (Magh-Cit), used as control, was evaluated by tumor volume reduction, histology, and morphometric analysis. Magh-Rh2Cit and Magh-Cit promoted a significant decrease in tumor area, and no experimental groups presented hematotoxic effects or increased levels of serum ALT and creatinine. This observation was corroborated by the histopathological examination of the liver and kidney of mice. Furthermore, the presence of nanoparticles was verified in lung tissue with no morphological changes, supporting the idea that our nanoformulations did not induce toxicity effects. No studies about the systemic action of rhodium (II) citrate-loaded maghemite nanoparticles have been carried out, making this report a suitable starting point for exploring the therapeutic potential of these compounds in treating breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Férricos/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Rodio/farmacología , Alanina Transaminasa/sangre , Animales , Femenino , Compuestos Férricos/toxicidad , Hepatocitos/patología , Riñón/fisiopatología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Rodio/toxicidad , Tasa de Supervivencia
19.
Breast Cancer Res Treat ; 147(3): 501-12, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25200444

RESUMEN

Breast cancer drug development costs nearly $610 million and 37 months in preclinical mouse model trials with minimal success rates. Despite these inefficiencies, there are still no consensus breast cancer preclinical models. Murine mammary adenocarcinoma 4T1-luc2 cells were implanted subcutaneous (SQ) or orthotopically percutaneous (OP) injection in the area of the nipple, or surgically into the chest 2nd mammary fat pad under direct vision (ODV) in Balb/c immunocompetent mice. Tumor progression was followed by in vivo bioluminescence and direct measurements, pathology and survival determined, and tumor gene expression analyzed by genome-wide microarrays. ODV produced less variable-sized tumors and was a reliable method of implantation. ODV implantation into the chest 2nd mammary pad rather than into the abdominal 4th mammary pad, the most common implantation site, better mimicked human breast cancer progression pattern, which correlated with bioluminescent tumor burden and survival. Compared to SQ, ODV produced tumors that differentially expressed genes whose interaction networks are of importance in cancer research. qPCR validation of 10 specific target genes of interest in ongoing clinical trials demonstrated significant differences in expression. ODV implantation into the chest 2nd mammary pad provides the most reliable model that mimics human breast cancer compared from subcutaneous implantation that produces tumors with different genome expression profiles of clinical significance. Increased understanding of the limitations of the different preclinical models in use will help guide new investigations and may improve the efficiency of breast cancer drug development .


Asunto(s)
Neoplasias de la Mama/patología , Trasplante de Células/métodos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Mediciones Luminiscentes , Glándulas Mamarias Animales/cirugía , Neoplasias Mamarias Experimentales/mortalidad , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante Heterólogo/métodos , Carga Tumoral
20.
Cancer Chemother Pharmacol ; 74(3): 559-69, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25053391

RESUMEN

PURPOSE: This study was undertaken to assess the possible modulatory effects and mechanisms of meloxicam, a cyclooxygenase-2 inhibitor, on the antitumor activity and cardiotoxicity of doxorubicin in a mice model of mammary carcinoma. METHODS: Solid tumor mass was developed in female albino mice using Ehrlich carcinoma cells. Forty mice-bearing tumor were divided randomly into four groups for treatment: with saline, meloxicam 10 mg/kg, doxorubicin 5 mg/kg and meloxicam 1 h ahead of doxorubicin, twice weekly for 2 weeks. Tumor volume was followed up and cardiac protective utility was estimated via measuring heart and serum parameters. RESULTS: Meloxicam expressed a non-significant increase in doxorubicin antitumor activity. Conversely, meloxicam significantly (p < 0.01) mitigated doxorubicin-induced elevation of serum cardiac enzymes [creatine kinase, lactate dehydrogenase and troponin-I]; cardiac lipid peroxidations marker; cardiac active caspase-3 content; and cardiac prostaglandin E2 content. Meloxicam significantly abrogated doxorubicin-induced disturbance in heart histology and relative heart weight to body weight. Meloxicam normalized doxorubicin-induced suppression in heart antioxidant enzymes activities and gene expressions [superoxide dismutase, glutathione peroxidase (GSH-Px) and catalase], and heart GSH content. In addition, meloxicam ameliorated doxorubicin-induced disturbance in phase II metabolizing enzyme, cardiac quinone reductase (QR), at activity level and mRNA expression. CONCLUSION: Meloxicam protects heart against doxorubicin toxicity without affecting its antitumor activity against solid mammary cancer model in mice. This protective effect is attributed to antioxidant effect, antiradical effect, antiinflammatory action, antiapoptotic effect and induction of QR enzyme.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Tiazinas/farmacología , Tiazoles/farmacología , Animales , Antibióticos Antineoplásicos/efectos adversos , Caspasa 3/metabolismo , Catalasa/genética , Catalasa/metabolismo , Dinoprostona/metabolismo , Femenino , Glutatión/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Neoplasias Mamarias Experimentales/mortalidad , Neoplasias Mamarias Experimentales/patología , Meloxicam , Ratones , Miocardio/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...