Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Clin Oncol ; 29(10): 1516-1527, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39017806

RESUMEN

BACKGROUND: In the THOR trial (NCT03390504) Cohort 1, erdafitinib demonstrated significantly prolonged overall survival (OS) (median 12.1 versus 7.8 months) and reduced risk of death by 36% (hazard ratio 0.64, P = 0.005) compared with chemotherapy in metastatic urothelial carcinoma (mUC) patients with FGFR alterations who progressed after ≥ 1 prior treatments, including anti-PD-(L)1. There have been no reports of the Japanese subgroup results yet. METHODS: THOR Cohort 1 randomized patients to erdafitinib once daily or docetaxel/vinflunine once every 3 weeks. Primary endpoint was OS. Secondary endpoints included progression-free survival (PFS) and objective response rate (ORR). No specific statistical power was set for this Japanese subgroup analysis. RESULTS: Of 266 patients randomized, 27 (14 erdafitinib; 13 chemotherapy) were Japanese. Baseline characteristics were generally similar between treatments and to the overall population, except for more males, lower body weight, and more upper tract primary tumors among Japanese patients. Compared with chemotherapy, erdafitinib showed improved OS (median 25.4 versus 12.4 months), PFS (median 8.4 versus 2.9 months) and ORR (57.1% versus 15.4%). Any grade treatment-related adverse events (AEs) occurred in all patients from both arms but Grade 3/4 AEs and AEs leading to discontinuation were lower in the erdafitinib arm. No new safety signals were observed in the Japanese subgroup. CONCLUSION: In the Japanese subgroup, erdafitinib showed improved survival and response compared to chemotherapy, with no new safety concerns. These results support erdafitinib as a treatment option for Japanese mUC patients with FGFR alterations, and early FGFR testing after diagnosis of mUC should be considered.


Asunto(s)
Quinoxalinas , Humanos , Masculino , Femenino , Anciano , Persona de Mediana Edad , Quinoxalinas/uso terapéutico , Anciano de 80 o más Años , Pirazoles/uso terapéutico , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/patología , Receptores de Factores de Crecimiento de Fibroblastos , Japón , Supervivencia sin Progresión , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Transicionales/tratamiento farmacológico , Carcinoma de Células Transicionales/secundario , Adulto , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología , Mutación , Pueblos del Este de Asia
2.
N Engl J Med ; 389(21): 1961-1971, 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37870920

RESUMEN

BACKGROUND: Erdafitinib is a pan-fibroblast growth factor receptor (FGFR) inhibitor approved for the treatment of locally advanced or metastatic urothelial carcinoma in adults with susceptible FGFR3/2 alterations who have progression after platinum-containing chemotherapy. The effects of erdafitinib in patients with FGFR-altered metastatic urothelial carcinoma who have progression during or after treatment with checkpoint inhibitors (anti-programmed cell death protein 1 [PD-1] or anti-programmed death ligand 1 [PD-L1] agents) are unclear. METHODS: We conducted a global phase 3 trial of erdafitinib as compared with chemotherapy in patients with metastatic urothelial carcinoma with susceptible FGFR3/2 alterations who had progression after one or two previous treatments that included an anti-PD-1 or anti-PD-L1. Patients were randomly assigned in a 1:1 ratio to receive erdafitinib or the investigator's choice of chemotherapy (docetaxel or vinflunine). The primary end point was overall survival. RESULTS: A total of 266 patients underwent randomization: 136 to the erdafitinib group and 130 to the chemotherapy group. The median follow-up was 15.9 months. The median overall survival was significantly longer with erdafitinib than with chemotherapy (12.1 months vs. 7.8 months; hazard ratio for death, 0.64; 95% confidence interval [CI], 0.47 to 0.88; P = 0.005). The median progression-free survival was also longer with erdafitinib than with chemotherapy (5.6 months vs. 2.7 months; hazard ratio for progression or death, 0.58; 95% CI, 0.44 to 0.78; P<0.001). The incidence of grade 3 or 4 treatment-related adverse events was similar in the two groups (45.9% in the erdafitinib group and 46.4% in the chemotherapy group). Treatment-related adverse events that led to death were less common with erdafitinib than with chemotherapy (in 0.7% vs. 5.4% of patients). CONCLUSIONS: Erdafitinib therapy resulted in significantly longer overall survival than chemotherapy among patients with metastatic urothelial carcinoma and FGFR alterations after previous anti-PD-1 or anti-PD-L1 treatment. (Funded by Janssen Research and Development; THOR ClinicalTrials.gov number, NCT03390504.).


Asunto(s)
Anticuerpos Monoclonales Humanizados , Antineoplásicos , Carcinoma de Células Transicionales , Receptores de Factores de Crecimiento de Fibroblastos , Neoplasias de la Vejiga Urinaria , Adulto , Humanos , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Transicionales/tratamiento farmacológico , Carcinoma de Células Transicionales/mortalidad , Carcinoma de Células Transicionales/patología , Docetaxel/efectos adversos , Docetaxel/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico
3.
Drug Alcohol Depend ; 233: 109331, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35149439

RESUMEN

BACKGROUND: Opioid use during pregnancy has been associated with adverse maternal and infant health outcomes. Prescription drug monitoring programs (PDMP) provide a population-based source of prescription data. We linked statewide PDMP and birth certificate data in Tennessee (TN) to determine patterns of prescription opioid and benzodiazepine use during pregnancy. METHODS: We constructed a cohort of 311,217 live singleton births from 2013 to 2016 with prescription history from 90 days before pregnancy to birth. Descriptive statistics were used to describe opioid prescription patterns during pregnancy overall, by maternal characteristics and by year. Multivariable logistic regression models estimated adjusted odds ratios and 95% confidence intervals for factors associated with prescription use. RESULTS: The prevalence of prescription use during pregnancy was 14.1% for opioid analgesics, 1.6% buprenorphine for medication-assisted treatment, and 2.6% for benzodiazepines. The prevalence of opioid analgesic use decreased from 16.6% (2013) to 11.8% (2016) (ptrend< 0.001). About 25% used for > 7 and 9.7% for > 30 days' supply. The most common types were hydrocodone (9.3%), codeine (3.4%), and oxycodone (2.9%). In adjusted models, lower education, lower income, pre-pregnancy obesity and smoking during pregnancy were associated with increased odds of any opioid and opioid analgesic use. CONCLUSION(S): Despite the encouraging trend of decreasing use of prescription opioid analgesics, the overall prevalence remained close to 12% with many women using for long durations. Use was associated with lower socioeconomic status, obesity, and prenatal smoking. Findings highlight the need for maternal education and resources, and provider support for implementation of evidence-based care.


Asunto(s)
Trastornos Relacionados con Opioides , Programas de Monitoreo de Medicamentos Recetados , Analgésicos/uso terapéutico , Analgésicos Opioides/uso terapéutico , Benzodiazepinas/uso terapéutico , Estudios de Cohortes , Femenino , Humanos , Obesidad , Trastornos Relacionados con Opioides/tratamiento farmacológico , Trastornos Relacionados con Opioides/epidemiología , Pautas de la Práctica en Medicina , Embarazo
4.
Blood ; 122(14): 2331-7, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23950178

RESUMEN

Panobinostat is an oral pan-deacetylase inhibitor that synergizes with bortezomib to inhibit both the aggresome and proteasome pathways in preclinical studies. PANORAMA 2 is a phase 2 trial of panobinostat in combination with bortezomib and dexamethasone to treat patients with relapsed and bortezomib-refractory multiple myeloma (with ≥2 prior lines of therapy, including an immunomodulatory drug, and patients who had progressed on or within 60 days of the last bortezomib-based therapy). Fifty-five heavily pretreated patients were enrolled (median, 4 prior regimens, including a median of 2 prior bortezomib-containing regimens). The overall response rate was 34.5% (1 near-complete response and 18 partial responses). An additional 10 patients achieved minimal response, for a clinical benefit rate of 52.7%. Median exposure and progression-free survival were 4.6 and 5.4 months, respectively. In patients who achieved a response, median time to response was 1.4 months, and median duration of response was 6.0 months. Common grade 3/4 adverse events, regardless of study drug relationship, included thrombocytopenia (63.6%), fatigue (20.0%), and diarrhea (20.0%). Only 1 patient had grade 3 peripheral neuropathy. Panobinostat, when combined with bortezomib and dexamethasone, can recapture responses in heavily pretreated, bortezomib-refractory multiple myeloma patients. This trial was registered at www.clinicaltrials.gov as #NCT01083602.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/efectos adversos , Bortezomib , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/efectos adversos , Indoles/administración & dosificación , Indoles/efectos adversos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Recurrencia Local de Neoplasia/mortalidad , Panobinostat , Pirazinas/administración & dosificación , Pirazinas/efectos adversos
5.
Free Radic Biol Med ; 51(3): 619-25, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21605664

RESUMEN

Emerging research suggests that antioxidant gene expression has the potential to suppress the development of gastroparesis. However, direct genetic evidence that definitively supports this concept is lacking. We used mice carrying a targeted disruption of Nfe2l2, the gene that encodes the transcription factor NRF2 and directs antioxidant Phase II gene expression, as well as mice with a targeted disruption of Gclm, the modifier subunit for glutamate-cysteine ligase, to test the hypothesis that defective antioxidant gene expression contributes to development of gastroparesis. Although expression of heme oxygenase-1 remained unchanged, expression of GCLC, GCLM, SOD1, and CAT was down-regulated in gastric tissue from Nrf2(-/-) mice compared to wild-type animals. Tetrahydrobiopterin oxidation was significantly elevated and nitrergic relaxation was impaired in Nrf2(-/-) mouse gastric tissue. In vitro studies showed a significant decrease in NO release in Nrf2(-/-) mouse gastric tissue. Nrf2(-/-) mice displayed delayed gastric emptying. The use of Gclm(-/-) mice demonstrated that the loss of glutamate-cysteine ligase function enhanced tetrahydrobiopterin oxidation while impairing nitrergic relaxation. These results provide genetic evidence that loss of antioxidant gene expression can contribute to the development of gastroparesis and suggest that NRF2 represents a potential therapeutic target.


Asunto(s)
Mucosa Gástrica/metabolismo , Gastroparesia/genética , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas Nitrérgicas/patología , Estómago/irrigación sanguínea , Animales , Antioxidantes/metabolismo , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Vaciamiento Gástrico/genética , Gastroparesia/patología , Gastroparesia/fisiopatología , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Humanos , Ratones , Ratones Noqueados , Relajación Muscular/genética , Factor 2 Relacionado con NF-E2/genética , Estómago/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Sistema Vasomotor
6.
J Biochem Mol Toxicol ; 24(3): 187-94, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20583300

RESUMEN

We reported earlier in a guinea pig model that exposure of 2-chloroethyl ethyl sulfide (CEES), a mustard gas analog, causes lung injury associated with the activation of tumor necrosis factor alpha (TNF-alpha), mitogen activated protein kinases (MAPK) signaling, and activator protein-1 (AP-1) transcription factor. Our earlier studies also revealed that antioxidant liposomes can be used as antidotes. Proinflammatory cytokines IL-1, IL-6, and TNF-alpha, either alone or in combination, can induce the activation of another group of transcription factors, namely SAF-1 (serum accelerator factor-1)/MAZ (Myc-associated zinc finger protein). Phosphorylation of SAF-1 via MAPK markedly increases its DNA-binding and transactivational potential. The objective of the present study was to investigate whether CEES exposure causes activation of IL-1 beta, IL-6, and SAF-1/MAZ and whether these effects can be prevented by antioxidant liposomes. A single dose (200 microL) of the antioxidant liposome mixture was administered intratracheally after 5 min of exposure of CEES (0.5 mg/kg). The animals were sacrificed either 1 h or 30 days after CEES exposure. CEES exposure caused an upregulation of proinflammatory cytokines IL-6 and IL-1 beta in the lung along with an increase in the activation of transcription factor SAF-1/MAZ. The antioxidant liposomes treatment significantly blocked the CEES-induced activation of IL-6, IL-1 beta, and SAF-1/MAZ. This might suggest that antioxidant liposomes might offer a potential therapeutic strategy against inflammatory diseases associated with activation of these bioactive molecules.


Asunto(s)
Antioxidantes/farmacología , Proteínas de Unión al ADN/fisiología , Inflamación/prevención & control , Liposomas/farmacología , Pulmón/efectos de los fármacos , Gas Mostaza/análogos & derivados , Factores de Transcripción/fisiología , Animales , Proteínas de Unión al ADN/análisis , Cobayas , Interleucina-1beta/biosíntesis , Interleucina-6/biosíntesis , Pulmón/patología , Masculino , Gas Mostaza/toxicidad , Factores de Transcripción/análisis
7.
Mol Cell Biochem ; 340(1-2): 203-13, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20204676

RESUMEN

This study aims to establish the antiproliferative effects of PK11195, a peripheral benzodiazepine receptor antagonist (PBR) in rat mammary tumor cells. Breast tumors were induced by administration of a carcinogen, dimethylbenz[a]anthracene to 50-day-old female rats maintained on a standard AIN-76A diet with casein as the protein source. The tumors were developed approximately after 120 days. The tumors were of grade I (20%), grade II (60%), and grade III (20%). The tumors were isolated and cultured in DMEM/F12 media with supplements. We characterized the properties of the isolated cells and study the effect of PK11195 on those cells. We were successful in growing breast tumor cells up to 30 passages for cellular characterization. These cells had high reactivity with Ki-67 and PCNA antibodies suggesting high proliferation rate. These cells were highly invasive as evident by matrigel invading ability. Furthermore, these cells acquired a positive response for CD-31 and VEGF antibodies suggesting angiogenic potential, and also possessed migrating ability/motility as evident by the wound healing properties. These cells expressed elevated levels of PBR, a cancer promoting gene. The proliferation, invasion and migration appear to decrease when treated with PK11195, a PBR antagonist. Furthermore, PK11195 treatment caused an increase in apoptosis as evident by increase in the levels of annexin V. However, the inhibition of cell proliferation by PK11195 was counteracted by Ro5-4864, a PBR agonist. Thus, PBR antagonist may be a potential therapeutic agent for the control of aggressiveness of breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Portadoras/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Antagonistas de Receptores de GABA-A , Isoquinolinas/farmacología , Neoplasias Mamarias Experimentales/patología , 9,10-Dimetil-1,2-benzantraceno , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Benzodiazepinonas/farmacología , Proteínas Portadoras/agonistas , Proteínas Portadoras/metabolismo , Ciclo Celular/efectos de los fármacos , Movimiento Celular , Relación Dosis-Respuesta a Droga , Femenino , Agonistas de Receptores de GABA-A , Antígeno Ki-67/metabolismo , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/metabolismo , Invasividad Neoplásica , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Toxicology ; 261(3): 143-51, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19464336

RESUMEN

We have recently reported that antioxidant liposomes can be used as antidotes for mustard gas induced lung injury in guinea pigs. The maximum protection was achieved with a liposome composed of tocopherols (alpha, gamma, delta) and N-acetylcysteine (NAC) when administered after 5 min of exposure of 2-chloroethyl ethyl sulfide (CEES), a half sulfur mustard gas. We also reported an association of mustard gas-induced lung injury with an activation of MAPK/AP-1 signaling pathway and cell proliferation. The objective of the present study was to investigate whether CEES-induced MAPKs/AP-1 signaling pathway is influenced by antioxidant liposome therapy. A single dose (200 microl) of the antioxidant liposome was administered intratracheally after 5 min of exposure of CEES (0.5 mg/kg). The animals were sacrificed after 1h and 30 days of CEES exposure. Although the liposome treatment did not have any significant effect on the activation of the MAPKs family (ERK1/2, p38 and JNK1/2), it significantly counteracted the CEES-induced activation of AP-1 transcription factors and corresponding increase in the protein levels of Fos, ATF and Jun family members. The liposome treatment significantly blocked the CEES-induced increase in the protein levels of cyclin D1, a cell cycle protein and PCNA, a cell differentiation marker. Furthermore, it protected lung against CEES-induced inflammation and infiltration of neutrophils, eosinophils and erythrocytes in the alveolar space. This suggests that the protective effect of antioxidant liposome against CEES-induced lung damage is mediated via control of AP-1 signaling.


Asunto(s)
Acetilcisteína/farmacología , Antídotos/farmacología , Antioxidantes/farmacología , Lesión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Tocoferoles/farmacología , Factor de Transcripción AP-1/metabolismo , Acetilcisteína/administración & dosificación , Factores de Transcripción Activadores/metabolismo , Animales , Antídotos/administración & dosificación , Antioxidantes/administración & dosificación , Proteínas Sanguíneas/metabolismo , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Eritrocitos/efectos de los fármacos , Cobayas , Liposomas , Pulmón/enzimología , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/enzimología , Lesión Pulmonar/patología , Masculino , Gas Mostaza/análogos & derivados , Infiltración Neutrófila/efectos de los fármacos , Fosforilación , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Eosinofilia Pulmonar/inducido químicamente , Eosinofilia Pulmonar/prevención & control , Factores de Tiempo , Tocoferoles/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
9.
Life Sci ; 83(19-20): 661-5, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18854196

RESUMEN

AIM: The present study aims to establish that cholinephosphotransferase (CPT), the terminal enzyme for the de novo biosynthesis of phosphatidylcholine (PC), can be used as a biomarker for breast cancer in an animal model. MAIN METHODS: Breast cancer was induced by intragastric administration of dimethylbenz(a)anthracene (DMBA) in rats. The activity and expression of CPT were compared between normal breast tissues and breast tumors. To establish possible mechanistic model, we looked into other enzymes of PC biosynthesis as well as c-fos protein expression and DNA binding. KEY FINDINGS: CPT enzyme activity and its expression were significantly higher in breast cancer tissues relative to normal breast tissues. Corresponding to the increase in the CPT activity and its expression, c-fos activity and its expression were also increased in breast tumors. SIGNIFICANCE: The present study suggests that increased CPT activity and expression is associated with DMBA-induced breast cancer development.


Asunto(s)
Diacilglicerol Colinafosfotransferasa/biosíntesis , Diacilglicerol Colinafosfotransferasa/genética , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Actinas/biosíntesis , Actinas/genética , Animales , Biomarcadores de Tumor , Western Blotting , Carcinógenos/toxicidad , Citidina Difosfato Colina/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Toxicol Lett ; 181(2): 112-7, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18675330

RESUMEN

We reported earlier that the activation of free-radical-mediated tumor necrosis factor-alpha (TNF-alpha) cascade is the major pathway in the inflammatory lung disease induced by 2-chloroethyl ethyl sulfide (CEES), a mustard gas analog. TNF-alpha induces activating protein 1 (AP-1) activation via phosphorylation of mitogen activated protein kinases (MAPKs). The present study examines the relationship between CEES induced lung injury and MAPKs signaling pathway. Adult guinea pigs received single intratracheal injection of different doses of CEES and were sacrificed at different time points. CEES exposure caused lung injury with evidence of fibrosis. The optimum activation of all members of the MAPKs family (ERK1/2, p38 and JNK1/2) was achieved at 0.5 mg/kg dose and at 1h. No significant change was observed beyond that time point. This led to an activation of AP-1 transcription factors associated with an increase in the protein levels of Fos, activating transcription factor (ATF) and Jun family members. To explore the involvement of AP-1 in cell proliferation, we determined the protein levels of cell cycle protein cyclin D1 and cell differentiation marker proliferating cell nuclear antigen (PCNA). An up regulation of these proteins was observed. Hence it is suggested that CEES exposure causes accumulation of TNF-alpha, which is associated with an activation of MAPK/AP-1 signaling pathway and cell proliferation. Further studies are needed to clarify whether the observed effects are the adaptive responses of the lung or they contribute to the lung injury.


Asunto(s)
Pulmón/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Gas Mostaza/análogos & derivados , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/fisiología , Animales , Western Blotting , Ciclina D1/análisis , ADN/metabolismo , Cobayas , Masculino , Gas Mostaza/toxicidad , Antígeno Nuclear de Célula en Proliferación/análisis , Factor de Necrosis Tumoral alfa/biosíntesis
11.
Mol Carcinog ; 47(4): 310-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17932947

RESUMEN

Soy protein is known to have breast tumor suppressing activity. The expression of peripheral benzodiazepine receptors (PBRs), currently renamed as translocator protein (TSPO) and their associated functions, such as nuclear cholesterol uptake and content also have been shown to be increased in breast cancer. Here we investigated whether the breast tumor suppressing effects of soy protein is mediated by down-regulation of PBR expression and function. Breast tumors were induced by gavage administration of a single dose (80 mg/kg) of dimethylbenz[a]anthracene (DMBA) into 50-d old female Sprague Dawley rats, maintained on a standard AIN-76A diet containing either casein or soy protein. Approximately 120 d following DMBA administration, the animals were sacrificed. All tumors were detected by palpation and at autopsy biopsy specimens were taken for histological grading. The ligand binding capacity, expression, and protein levels of PBRs, their nuclear localization and function, such as nuclear cholesterol uptake and content, were significantly increased in the tumors. However, replacement of casein by soy protein in the diet caused a significant decrease in all of these parameters. These data suggest that soy protein inhibits breast tumor development by decreasing the expression of the tumor-promoting gene, which encodes PBRs.


Asunto(s)
Neoplasias Mamarias Experimentales/dietoterapia , Receptores de GABA-A/fisiología , Proteínas de Soja/uso terapéutico , 9,10-Dimetil-1,2-benzantraceno/farmacología , Animales , Benzodiazepinonas/metabolismo , Carcinógenos/farmacología , Femenino , Ligandos , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Proteínas de Soja/administración & dosificación , Tritio/metabolismo
12.
Mol Cell Biochem ; 290(1-2): 169-76, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16941229

RESUMEN

This study was to demonstrate by histological grading whether soy protein protects against dimethylbenz[a]anthracene (DMBA) -induced breast tumors in female rats. At 25 days of age, rats were fed diets containing either casein or soy protein. After 25 days on diets, a single dose of DMBA in sesame oil (80 mg/kg) was administered by gavage. All tumors were detected by palpation. The number of tumors per rat was less in soy group than that in casein group at any time point up to 122 days after DMBA administration. Incidence of tumors was less in soy protein group than that in casein group. Casein group had 20% grade I, 60% grade II, and 20% grade III adenocarcinoma. However, the soy group had 100% grade I adenocarcinoma and no aggressive grade II or grade III tumor. There was a delay in the development of tumor in the soy protein group in comparison to the casein group. Again, unlike casein, the soy group had cessation of angiogenesis at several sites of tumor, and reduced levels of angiogenic markers, VEGF and bFGF. Immunohistochemical analysis of the breast tissues did not show any CD-31 positive stain in soy protein group, whereas some CD-31 positive stain was revealed in casein group, which further suggests that soy protein controls angiogenesis. Furthermore, proliferative index as assessed by Ki-67 staining was less in soy protein group than that in casein group. These findings suggest that the soy protein may protect against the development of a more aggressive breast carcinoma.


Asunto(s)
Adenocarcinoma/prevención & control , Antracenos , Proteínas en la Dieta/administración & dosificación , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/terapia , Proteínas de Soja/administración & dosificación , Adenocarcinoma/patología , Adenocarcinoma/ultraestructura , Animales , Western Blotting , Mama/efectos de los fármacos , Mama/ultraestructura , Femenino , Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Mamarias Experimentales/ultraestructura , Estadificación de Neoplasias , Ratas , Ratas Sprague-Dawley
13.
Glycoconj J ; 23(3-4): 199-207, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16691503

RESUMEN

Expression of peripheral benzodiazepine receptors (PBR) has been found in every tissue examined; however, it is most abundant in steroid-producing tissues. Although the primary function of PBR is the regulation of steroidogenesis, its existence in nonsteroidogenic tissues as well as in other cellular compartments including the nucleus suggests that there may be other roles for PBR. Our laboratory reported earlier a significant increase of PBR density in the nucleus of DMBA-induced malignant submandibular glands of rats, suggesting a role of PBR in nuclear events of peripheral tissues. Since then numerous studies have demonstrated the abundance of PBR in tumors. Numerous studies implicate a role for cholesterol in the mechanisms underlying cell proliferation and cancer progression. Based on studies with a battery of human breast cancer cell lines and several human tissue biopsies, Hardwick et al. suggested that PBR expression, nuclear localization, and PBR-mediated cholesterol transport into the nucleus are involved in human breast cancer cell proliferation and aggressive phenotype expression. The purpose of the present study is to confirm this hypothesis by developing an animal breast cancer model and correlating the above events with the breast cancer. Weanling rats were maintained on a diet containing animal protein (casein) for 30 days and then a single dose of DMBA in sesame oil (80 mg/kg) was administered by gavage to the animals. Control animals received the vehicle only. After 122 days of DMBA administration, the animals were sacrificed. All tumors were detected by palpation. B(max) of PBRs was 52.6% and 128.4% higher in the non-aggressive and aggressive cancer tissues, respectively, than that in normal tissues. Cholesterol uptake into isolated nuclei was found to be higher in both non-aggressive and aggressive tumor breast tissue than that in control tissue. There was also corresponding increase in B(max) of PBRs in the nucleus of cancer tissues. Furthermore, the nuclear nucleoside triphosphatase (NTPase) activity was found to be higher in aggressive tumor tissues than that in non-aggressive tumor tissues. In conclusion, these data suggest that PBR ligand binding, and PBR-mediated cholesterol transport into the nucleus may be involved in the development of mammary gland adenocarcinoma, thus participating in the advancement of the disease.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/toxicidad , Proteínas Portadoras/metabolismo , Neoplasias Mamarias Animales/metabolismo , Receptores de GABA-A/metabolismo , Animales , Benzodiazepinonas/metabolismo , Transporte Biológico , Western Blotting , Carcinógenos/toxicidad , Núcleo Celular/metabolismo , Colesterol/metabolismo , Femenino , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/patología , Nucleósido-Trifosfatasa/metabolismo , Ratas , Ratas Sprague-Dawley
14.
J Biomed Biotechnol ; 2004(4): 195-202, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15467159

RESUMEN

Manganese superoxide dismutase (Mn-SOD), localized at the mitochondrial matrix, has the ability to protect cells against oxidative damage. It has been reported that low levels of Mn-SOD gene expression cause the development of certain kind of tumors. On the other hand, overexpression of Mn-SOD gene may play an important role in the development of cancer. In our study, we find that Mn-SOD activity was higher in nonaggressive (MCF-7) and aggressive (BT-549 and 11-9-14) breast cancer cell lines compared to that of nontumorigenic (MCF-12A and MCF-12F) mammary epithelial cell lines. We also observed an increased expression of Mn-SOD gene in cancerous cell lines. The elevated level of SOD activity in nonaggressive and aggressive breast epithelial cell lines was associated with some changes in nucleotide sequence.

15.
Mol Cancer Ther ; 3(2): 199-204, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14985460

RESUMEN

Cadmium (Cd) is an ubiquitous environmental carcinogen. Membrane phospholipids as well as fatty acid profile of membrane phospholipids are known to be altered in tumorigenicity and malignancy. Synthesis of cellular phosphatidylcholine (PC) has been used as a marker for membrane proliferation in the neoplastic mammary gland tissue. Cholinephosphotransferase (CPT), the terminal enzyme in de novo synthesis of PC, has an important role in regulating the acyl group of PC in mammalian cells. Our previous studies have shown that CPT is expressed differentially in the normal and cancerous mammary epithelial cell lines. In this study, we examined the effect of cadmium on CPT activity using normal (MCF-12A and MCF-12F) and cancerous (MCF-7, BT-549, and 11-9-1-4) human mammary epithelial cell lines. There was no consistent pattern of CPT activity in response to different doses of cadmium. The activity did not show a time-dependent variation at 5 micro M concentration, except in MCF-7 and 11-9-1-4. CPT gene expression increased with cadmium as evident from slot blots. Mutation in the nucleotide sequence was also observed as the result of cadmium but this did not result into amino acid sequence changes.


Asunto(s)
Neoplasias de la Mama/enzimología , Cadmio/farmacología , Carcinógenos/farmacología , Diacilglicerol Colinafosfotransferasa/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Glándulas Mamarias Humanas/enzimología , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Tamaño de la Célula , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/patología , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/efectos de los fármacos , Mutagénesis/efectos de los fármacos
16.
Indian J Exp Biol ; 41(4): 283-9, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15255635

RESUMEN

Chronic ingestion (for 22-30 consecutive days) of caffeine (20 mg/kg/day, p.o.) increased the activities of the hepatic enzymes- catalase (CAT) and superoxide dismutase (SOD) and decreased its lipid peroxidation (LP) in mice. Development of Ehrlich ascites carcinoma (EAC) cell decreased the activities of hepatic CAT and SOD and increased LP. But pretreatment of caffeine for 12 consecutive days and continuation of its treatment during the course of development of EAC cells restored the EAC cell-induced changes in liver CAT, SOD and LP to their corresponding control values. Thus, the present results by confirming the results of others previously published, suggest that caffeine is an antioxidant and may act as an anticarcinogen.


Asunto(s)
Antioxidantes/metabolismo , Cafeína/farmacología , Carcinoma de Ehrlich/enzimología , Catalasa/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología , Hígado/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Animales , Cafeína/administración & dosificación , Carcinoma de Ehrlich/patología , Estimulantes del Sistema Nervioso Central/administración & dosificación , Femenino , Depuradores de Radicales Libres/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Hígado/enzimología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA