Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Curr Treat Options Oncol ; 25(6): 703-718, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38691257

RESUMO

OPINION STATEMENT: Dysphagia, difficulty in eating and drinking, remains the most common side effect of radiotherapy treatment for head and neck cancer (HNC) with devastating consequences for function and quality of life (QOL). Over the past decade, 5-year survival has improved due to multiple factors including treatment advances, reduction in smoking, introduction of the human papillomavirus (HPV) vaccine and more favourable prognosis of HPV-related cancers. Increased prevalence of HPV-positive disease, which tends to affect younger individuals, has led to an elevated number of people living for longer with the sequelae of cancer and its treatment. Symptoms are compounded by late effects of radiotherapy which may lead to worsening of dysphagia for some long-term survivors or new-onset dysphagia for others. Speech-language pathology (SLP) input remains core to the assessment and management of dysphagia following HNC treatment. In this article, we present current SLP management of dysphagia post-radiotherapy. We discuss conventional treatment approaches, the emergence of therapy adjuncts and current service delivery models. The impact of adherence on therapy outcomes is highlighted. Despite treatment advancements, patients continue to present with dysphagia which is resistant to existing intervention approaches. There is wide variation in treatment programmes, with a paucity of evidence to support optimal type, timing and intensity of treatment. We discuss the need for further research, including exploration of the impact of radiotherapy on the central nervous system (CNS), the link between sarcopenia and radiotherapy-induced dysphagia and the benefits of visual biofeedback in rehabilitation.


Assuntos
Transtornos de Deglutição , Neoplasias de Cabeça e Pescoço , Patologia da Fala e Linguagem , Humanos , Transtornos de Deglutição/etiologia , Transtornos de Deglutição/terapia , Patologia da Fala e Linguagem/métodos , Neoplasias de Cabeça e Pescoço/radioterapia , Neoplasias de Cabeça e Pescoço/terapia , Neoplasias de Cabeça e Pescoço/complicações , Qualidade de Vida , Gerenciamento Clínico , Radioterapia/efeitos adversos , Radioterapia/métodos
2.
Genesis ; 54(12): 626-635, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27731922

RESUMO

Temporal and spatial regulation of genes mediated by tissue-specific promoters and conditional gene expression systems provide a powerful tool to study gene function in health, disease, and during development. Although transgenic mice expressing the Cre recombinase in the gastric epithelium have been reported, there is a lack of models that allow inducible and reversible gene modification in the stomach. Here, we exploited the gastrointestinal epithelium-specific expression pattern of the three trefoil factor (Tff) genes and bacterial artificial chromosome transgenesis to generate a novel mouse strain that expresses the CreERT2 recombinase and the reverse tetracycline transactivator (rtTA). The Tg(Tff1-CreERT2;Tff2-rtTA;Tff3-Luc) strain confers tamoxifen-inducible irreversible somatic recombination and allows simultaneous doxycycline-dependent reversible gene activation in the gastric epithelium of developing and adult mice. This strain also confers luciferase activity to the intestinal epithelium to enable in vivo bioluminescence imaging. Using fluorescent reporters as conditional alleles, we show Tff1-CreERT2 and Tff2-rtTA transgene activity in a partially overlapping subset of long-term regenerating gastric stem/progenitor cells. Therefore, the Tg(Tff1-CreERT2;Tff2-rtTA;Tff3-Luc) strain can confer intermittent transgene expression to gastric epithelial cells that have undergone previous gene modification, and may be suitable to genetically model therapeutic intervention during development, tumorigenesis, and other genetically tractable diseases. Birth Defects Research (Part A) 106:626-635, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Mucosa Intestinal/metabolismo , Fator Trefoil-1/biossíntese , Fator Trefoil-2/biossíntese , Fator Trefoil-3/biossíntese , Animais , Regulação da Expressão Gênica no Desenvolvimento , Integrases/biossíntese , Integrases/genética , Mucosa Intestinal/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/genética , Recombinases/genética , Transgenes/genética , Fator Trefoil-1/genética , Fator Trefoil-2/genética , Fator Trefoil-3/genética
3.
Vet Immunol Immunopathol ; 181: 15-23, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26961672

RESUMO

There is strong evidence that high yielding dairy cows are extremely susceptible to infectious diseases, and that this has severe economic consequences for the dairy industry and welfare implications. Here we present preliminary functional evidence showing that the innate immune response differs between cow breeds. The ability of macrophages (MØ) to kill pathogens depends in part on oxygen-dependent and independent mechanisms. The oxygen-dependent mechanisms rely on the generation of reactive oxygen and nitrogen species (ROS/RNS, respectively). ROS production has been shown to activate the inflammasome complex in MØ leading to increased production of the pro-inflammatory cytokine Interleukin-1ß (IL-1ß). Conversely RNS inhibits inflammasome mediated IL-1ß activation, indicating a division between inflammasome activation and RNS production. In the present study MØ from Brown Swiss (BS) cattle produce significantly more RNS and less IL-1ß when compared to cells from Holstein Friesian (HF) cattle in response to bacterial or fungal stimuli. Furthermore, BS MØ killed ingested Salmonella typhimurium more efficiently, supporting anecdotal evidence of increased disease resistance of the breed. Inhibition of autophagy by 3-methyladenine (3-MA) stimulated IL-1ß secretion in cells from both breeds, but was more pronounced in HF MØ. Blocking RNS production by l-arginase completely abolished RNS production but increased IL-1ß secretion in BS MØ. Collectively these preliminary data suggest that the dichotomy of inflammasome activation and RNS production exists in cattle and differs between these two breeds. As pattern recognition receptors and signaling pathways are involved in the assessed functional differences presented herein, our data potentially aid the identification of in vitro predictors of appropriate innate immune response. Finally, these predictors may assist in the discovery of candidate genes conferring increased disease resistance for future use in combination with known production traits.


Assuntos
Bovinos/imunologia , Macrófagos/imunologia , Animais , Cruzamento , Imunidade Inata , Interleucina-18/biossíntese , Interleucina-1beta/biossíntese , Óxido Nítrico/biossíntese , Fagocitose , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
4.
Sci Signal ; 7(345): ra92, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25270258

RESUMO

Most colon cancers arise from somatic mutations in the tumor suppressor gene APC (adenomatous polyposis coli), and these mutations cause constitutive activation of the Wnt-to-ß-catenin pathway in the intestinal epithelium. Because Wnt-ß-catenin signaling is required for homeostasis and regeneration of the adult intestinal epithelium, therapeutic targeting of this pathway is challenging. We found that genetic activation of the cytokine-stimulated pathway mediated by the receptor gp130, the associated Jak (Janus kinase) kinases, and the transcription factor Stat3 (signal transducer and activator of transcription 3) was required for intestinal regeneration in response to irradiation-induced damage in wild-type mice and for tumorigenesis in Apc-mutant mice. Systemic pharmacological or partial genetic inhibition of gp130-Jak-Stat3 signaling suppressed intestinal regeneration, the growth of tumors in Apc-mutant mice, and the growth of colon cancer xenografts. The growth of Apc-mutant tumors depended on gp130-Jak-Stat3 signaling for induction of the polycomb repressor Bmi-1, and the associated repression of genes encoding the cell cycle inhibitors p16 and p21. However, suppression of gp130-Jak-Stat3 signaling did not affect Wnt-ß-catenin signaling or homeostasis in the intestine. Thus, these data not only suggest a molecular mechanism for how the gp130-Jak-Stat3 pathway can promote cancer but also provide a rationale for therapeutic inhibition of Jak in colon cancer.


Assuntos
Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes APC/fisiologia , Mucosa Intestinal/fisiologia , Regeneração/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Neoplasias do Colo/genética , Receptor gp130 de Citocina/metabolismo , Primers do DNA/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Técnicas Histológicas , Imuno-Histoquímica , Janus Quinase 1/metabolismo , Luciferases , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Via de Sinalização Wnt/genética
5.
Mol Cancer Ther ; 13(2): 468-74, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24398427

RESUMO

Aberrant activation of the latent transcription factor STAT3 and its downstream targets is a common feature of epithelial-derived human cancers, including those of the gastrointestinal tract. Mouse models of gastrointestinal malignancy implicate Stat3 as a key mediator of inflammatory-driven tumorigenesis, in which its cytokine/gp130/Janus kinase (Jak)-dependent activation provides a functional link through which the microenvironment sustains tumor promotion. Although therapeutic targeting of STAT3 is highly desirable, such molecules are not available for immediate clinical assessment. Here, we investigated whether the small-molecule Jak1/2 inhibitor AZD1480 confers therapeutic benefits in two mouse models of inflammation-associated gastrointestinal cancer, which are strictly dependent of excessive Stat3 activation. We confirm genetically that Cre-mediated, tumor cell-specific reduction of Stat3 expression arrests the growth of intestinal-type gastric tumors in gp130(F/F) mice. We find that systemic administration of AZD1480 readily replicates this effect, which is associated with reduced Stat3 activation and correlates with diminished tumor cell proliferation and increased apoptosis. Likewise, AZD1480 therapy also conferred a cytostatic effect on established tumors in a colitis-associated colon cancer model in wild-type mice. As predicted from our genetic observations in gp130(F/F) mice, the therapeutic effect of AZD1480 remains fully reversible upon cessation of compound administration. Collectively, our results provide the first evidence that pharmacologic targeting of excessively activated wild-type Jak kinases affords therapeutic suppression of inflammation-associated gastrointestinal cancers progression in vivo.


Assuntos
Neoplasias Gastrointestinais/prevenção & controle , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/prevenção & controle , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/metabolismo , Progressão da Doença , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética
6.
Oncol Rep ; 24(3): 779-85, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20664987

RESUMO

The anticancer effects elicited by epigallocatechin gallate (EGCG) are well established in various models of cancer, while raloxifene is as an established selective estrogen receptor modulator (SERM), which is not yet clinically utilized for the treatment of breast cancer. Previous study from this laboratory has demonstrated that the combination of EGCG (25 microM) and raloxifene (4 microM) elicits a strong cytotoxic response in MDA-MB-231 human breast cancer cells, which lack the estrogen receptor (ER) and erbB-2/ Her-2 receptor. This study was therefore designed to probe the mechanism underlying this cytotoxic response, with an emphasis on determining how the combination treatment influenced the total expression and phosphorylation of key signaling proteins. Specifically, following 12 and 18 h of the combination treatment, we observed significant decreases in the phosphorylation of the epidermal growth factor receptor (EGFR), AKT, mammalian target of rapamycin (mTOR) and S-6-kinase (S6K), and significant increases in the phosphorylation of stress activated protein kinases (SAPKs). Furthermore, these changes were associated with a reduction in the nuclear localization of p65, a major subunit of NF-kappaB. These results demonstrate that the combination of EGCG and raloxifene effectively reduced the mitogenic and survival signaling in MDA-MB-231 cells. Thus, this combination warrants further experimentation as a potential treatment for ER-negative breast cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Mama/patologia , Catequina/análogos & derivados , Catequina/farmacologia , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Receptor alfa de Estrogênio/deficiência , Feminino , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Cloridrato de Raloxifeno/farmacologia , Receptor ErbB-2/deficiência , Proteínas Quinases S6 Ribossômicas/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Fator de Transcrição RelA/metabolismo
7.
Cancer Epidemiol Biomarkers Prev ; 18(10): 2782-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19755648

RESUMO

Aberrant DNA methylation is a recognized feature of human cancers, and folate is directly involved in DNA methylation via one-carbon metabolism. Previous reports also suggest that folate status is associated with the natural history of human papillomavirus (HPV) infection. A cross-sectional study was conducted to test the hypothesis that folate status and aberrant DNA methylation show a progressive change across stages of cervical pathology from normal cells to cervical cancer. Additionally, we postulated that a gene-specific hypermethylation profile might be used as a predictive biomarker of cervical cancer risk. DNA hypermethylation of seven tumor suppressor genes, global DNA hypomethylation, systemic folate status, and HPV status were measured in 308 women with a diagnosis of normal cervix (n = 58), low-grade cervical intraepithelial neoplasia (CIN1; n = 68), high-grade cervical intraepithelial neoplasia (CIN2, n = 56; and CIN3, n = 76), or invasive cervical cancer (ICC; n = 50). Lower folate status was associated with high-risk HPV infection (P = 0.031) and with a diagnosis of cervical intraepithelial neoplasia or invasive cervical cancer (P < 0.05). Global DNA hypomethylation was greater in women with invasive cervical cancer than all other groups (P < 0.05). A cluster of three tumor suppressor genes, CDH1, DAPK, and HIC1, displayed a significantly increased frequency of promoter methylation with progressively more severe cervical neoplasia (P < 0.05). These findings are compatible with a role for folate in modulating the risk of cervical cancer, possibly through an influence over high-risk HPV infection. DAPK, CDH1, and HIC1 genes are potential biomarkers of cervical cancer risk.


Assuntos
Metilação de DNA , Ácido Fólico/metabolismo , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/metabolismo , Displasia do Colo do Útero/genética , Displasia do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas Reguladoras de Apoptose/genética , Caderinas/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Proteínas Quinases Associadas com Morte Celular , Eritrócitos/metabolismo , Feminino , Ácido Fólico/sangue , Glutationa S-Transferase pi/genética , Humanos , Pessoa de Meia-Idade , Infecções por Papillomavirus/patologia , Receptores do Ácido Retinoico/genética , Proteínas Supressoras de Tumor/genética , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/virologia , Adulto Jovem , Displasia do Colo do Útero/patologia , Displasia do Colo do Útero/virologia
8.
Life Sci ; 82(17-18): 943-8, 2008 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-18371987

RESUMO

Previous studies have demonstrated that raloxifene induces apoptosis in a variety of cancer cell lines. We aimed to determine if this effect was enhanced by combining raloxifene with epigallocatechin gallate (EGCG). Results demonstrated that EGCG (25 microM) and raloxifene (1-5 microM) produced enhanced cytotoxicity toward MDA-MB-231 breast cancer cells compared to either drug alone following 7 days of treatment. The combination of 5 microM raloxifene and EGCG was the most effective as it decreased cell number by 96% of control, and time-course studies demonstrated that significant cytotoxicity began 36 h after treatment. Potential mechanisms for this effect were then investigated. Flow cytometry experiments demonstrated that apoptosis was significantly increased following 12 h of combination treatment compared to all other treatment groups. A maximal increase in the proportion of cells in the G(1)-phase of the cell cycle (116% of control) occurred following 24 h of combination treatment, 12 h after the significant increase in apoptosis, and thus was not considered to be a viable mechanism for the enhancement of apoptosis. While raloxifene was a competitive inhibitor of microsomal UDP-glucuronosyltransferase activity (K(i) of 24 microM), it did not decrease the metabolism of EGCG as the rate of disappearance of EGCG from the media was the same for cells treated with either EGCG or EGCG+raloxifene. Finally, the combination treatment reduced the phosphorylation of EGFR and AKT proteins by 21.2+/-3.3% and 31.5+/-1.7% from control, respectively. In conclusion, the synergistic cytotoxicity elicited by the combination of EGCG and raloxifene results from an earlier and greater induction of apoptosis. This is likely to be a result of reduced phosphorylation of EGFR and AKT signaling proteins.


Assuntos
Apoptose/efeitos dos fármacos , Catequina/análogos & derivados , Fármacos Neuroprotetores/farmacologia , Cloridrato de Raloxifeno/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Western Blotting , Catequina/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Meios de Cultura , Receptores ErbB/metabolismo , Feminino , Citometria de Fluxo , Glucuronosiltransferase/metabolismo , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética
9.
Int J Cancer ; 122(9): 1966-71, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18098290

RESUMO

Both epigallocatechin gallate (EGCG) and curcumin have shown efficacy in various in vivo and in vitro models of cancer. This study was designed to determine the efficacy of these naturally derived polyphenolic compounds in vitro and in vivo, when given in combination. Studies in MDA-MB-231 cells demonstrated that EGCG + curcumin was synergistically cytotoxic and that this correlated with G(2)/M-phase cell cycle arrest. After 12 hr, EGCG (25 microM) + curcumin (3 microM) increased the proportion of cells in G(2)/M-phase to 263 +/- 16% of control and this correlated with a 50 +/- 4% decrease in cell number compared to control. To determine if this in vitro result would translate in vivo, athymic nude female mice were implanted with MDA-MB-231 cells and treated with curcumin (200 mg/kg/day, po), EGCG (25 mg/kg/day, ip), EGCG + curcumin, or vehicle control (5 ml/kg/day, po) for 10 weeks. Tumor volume in the EGCG + curcumin treated mice decreased 49% compared to vehicle control mice (p < 0.05), which correlated with a 78 +/- 6% decrease in levels of VEGFR-1 protein expression in the tumors. Curcumin treatment significantly decreased tumor protein levels of EGFR and Akt, however the expression of these proteins was not further decreased following combination treatment. Therefore, these results demonstrate that the combination of EGCG and curcumin is efficacious in both in vitro and in vivo models of ER alpha-breast cancer and that regulation of VEGFR-1 may play a key role in this effect.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biomarcadores Tumorais/análise , Neoplasias da Mama/tratamento farmacológico , Catequina/análogos & derivados , Curcumina/farmacologia , Receptor alfa de Estrogênio/análise , Animais , Western Blotting , Neoplasias da Mama/química , Catequina/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Proteína Oncogênica v-akt/metabolismo , Tamanho do Órgão , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Aumento de Peso
10.
Int J Oncol ; 30(6): 1407-12, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17487361

RESUMO

Potential mechanisms for the synergistic cytotoxicity elicited by epigallocatechin gallate (EGCG) (25 microM) and 4-hydroxytamoxifen (4-OHT) (1 microM) in MDA-MB-231 human breast cancer cells were investigated. The role of apoptosis was determined using chromatin condensation and Annexin-V staining. Condensed chromatin was visible following 24 h of combination treatment while flow cytometry experiments demonstrated that apoptosis was 2-fold greater following 36 h of combination treatment compared to EGCG. The temporal appearance of cells in G1-arrest did not correlate with apoptosis and thus was not considered to be a viable mechanism for the enhancement of apoptosis. While 4-OHT was a weak competitive inhibitor of microsomal UGT activity (Ki 95 microM), it did not alter the metabolism of EGCG as the rate of disappearance of EGCG from the media was the same for cells treated with either EGCG or EGCG + 4-OHT. Additionally, the metabolism of EGCG was not shifted toward the production of active methylated metabolites, as neither 4''-MeEGCG nor 4',4''-diMeEGCG (2.5-25 microM) were cytotoxic toward MDA-MB-231 cells. In conclusion, the synergistic cytotoxicity elicited by the combination of EGCG and 4-OHT results from an earlier induction of apoptosis but this was not caused by an increase in G1-arrest or 4-OHT-mediated changes in the metabolism of EGCG.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Catequina/análogos & derivados , Tamoxifeno/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Apoptose/efeitos dos fármacos , Catequina/metabolismo , Catequina/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Citometria de Fluxo , Humanos , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia
11.
Life Sci ; 79(25): 2329-36, 2006 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-16945390

RESUMO

Green tea and its major constituent epigallocatechin gallate (EGCG) have been extensively studied as a potential treatment for a variety of diseases, including cancer. Epidemiological data have suggested that EGCG may provide protective effects against hormone related cancers, namely breast or prostate cancer. Extensive in vitro investigations using both hormone responsive and non-responsive cell lines have shown that EGCG induces apoptosis and alters the expression of cell cycle regulatory proteins that are critical for cell survival and apoptosis. This review will highlight the important in vitro mechanistic actions elicited by EGCG in various breast and prostate cancer cell lines. Additionally, the actions of green tea/EGCG in in vivo models for these cancers as well as in clinical trials will be discussed.


Assuntos
Anticarcinógenos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Catequina/análogos & derivados , Neoplasias da Próstata/tratamento farmacológico , Apoptose/efeitos dos fármacos , Catequina/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Feminino , Humanos , Masculino , Chá/química , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA