Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Clin Cancer Res ; 27(10): 2706-2711, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33648989

RESUMO

The dramatic impact of the COVID-19 pandemic has resulted in an "all hands on deck" approach to find new therapies to improve outcomes in this disease. In addition to causing significant respiratory pathology, infection with SARS-CoV-2 (like infection with other respiratory viruses) directly or indirectly results in abnormal vasculature, which may contribute to hypoxemia. These vascular effects cause significant morbidity and may contribute to mortality from the disease. Given that abnormal vasculature and poor oxygenation are also hallmarks of solid tumors, lessons from the treatment of cancer may help identify drugs that can be repurposed to treat COVID-19. Although the mechanisms that result in vascular abnormalities in COVID-19 are not fully understood, it is possible that there is dysregulation of many of the same angiogenic and thrombotic pathways as seen in patients with cancer. Many anticancer therapeutics, including androgen deprivation therapy (ADT) and immune checkpoint blockers (ICB), result in vascular normalization in addition to their direct effects on tumor cells. Therefore, these therapies, which have been extensively explored in clinical trials of patients with cancer, may have beneficial effects on the vasculature of patients with COVID-19. Furthermore, these drugs may have additional effects on the disease course, as some ADTs may impact viral entry, and ICBs may accelerate T-cell-mediated viral clearance. These insights from the treatment of cancer may be leveraged to abrogate the vascular pathologies found in COVID-19 and other forms of hypoxemic respiratory failure.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Vasos Sanguíneos/efeitos dos fármacos , COVID-19/prevenção & controle , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , COVID-19/epidemiologia , COVID-19/virologia , Ensaios Clínicos como Assunto , Progressão da Doença , Humanos , Masculino , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Avaliação de Resultados em Cuidados de Saúde , Pandemias , Neoplasias da Próstata/irrigação sanguínea , Fatores de Risco , SARS-CoV-2/fisiologia
2.
Breast Cancer Res Treat ; 183(1): 145-151, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32607640

RESUMO

PURPOSE: A positive margin after breast conserving surgery has consistently been shown to be a significant predictor for ipsilateral breast tumor recurrence. Currently, there is no standard for intraoperative margin assessment during lumpectomy, and up to 20% of cases result in positive margins. MarginProbe is a device that provides real-time evaluation of lumpectomy margins during surgery. The aim of this study was to evaluate the impact of MarginProbe as an adjunct to standard operating procedure (SOP). METHODS: Patients diagnosed with breast cancer scheduled for breast conserving surgery were consented for intraoperative use of MarginProbe. Shaved margins were excised based on margin assessment using the surgeon's SOP which included specimen radiography and gross pathologic examination, and feedback from the device. The primary endpoint was re-excision rate. Secondary endpoints included sensitivity, specificity, false-positive and negative rates. RESULTS: Of the 60 breast cancers, initial histologically close/positive margins were identified in 18 patients (30%). The re-excision rate in the overall cohort was 6.6%, compared to a historical re-excision rate of 8.6% (p < 0.01). Based on 360 measurement sites, MarginProbe demonstrated a sensitivity of 67% and specificity of 60%, with a positive predictive value of 16%, and of negative predictive value of 94%, which was similar to the accuracy of SOP. CONCLUSIONS: MarginProbe performs equally as well as specimen radiography and gross pathologic examination. In this setting where the baseline re-excision rate was low, the use of MarginProbe as an adjunct to SOP resulted in a small 2% absolute reduction in re-excision rate.


Assuntos
Neoplasias da Mama/cirurgia , Carcinoma/cirurgia , Eletrodiagnóstico/instrumentação , Margens de Excisão , Mastectomia Segmentar/métodos , Recidiva Local de Neoplasia/prevenção & controle , Reoperação , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Carcinoma/irrigação sanguínea , Carcinoma/patologia , Núcleo Celular/fisiologia , Eletrodiagnóstico/métodos , Estrogênios , Feminino , Marcadores Fiduciais , Humanos , Cuidados Intraoperatórios/instrumentação , Potenciais da Membrana , Pessoa de Meia-Idade , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/patologia , Neoplasias Hormônio-Dependentes/cirurgia , Utilização de Procedimentos e Técnicas , Progesterona , Reoperação/estatística & dados numéricos
3.
Oncotarget ; 6(15): 13790-802, 2015 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-25912421

RESUMO

Fibroblast growth factor-8b (FGF8b) affects the epithelial/stromal compartments of steroid hormone-regulated tumors by exerting an autocrine activity on cancer cells and a paracrine pro-angiogenic function, thus contributing to tumor progression. The FGF8b/FGF receptor (FGFR) system may therefore represent a target for the treatment of steroid hormone-regulated tumors. The soluble pattern recognition receptor long pentraxin-3 (PTX3) binds various FGFs, including FGF2 and FGF8b, thus inhibiting the angiogenic and tumorigenic activity of androgen-regulated tumor cells. Nevertheless, the complex/proteinaceous structure of PTX3 hampers its pharmacological exploitation. In this context, the acetylated pentapeptide Ac-ARPCA-NH2 (ARPCA), corresponding to the N-terminal amino acid sequence PTX3(100-104), was identified as a minimal FGF2-binding peptide able to antagonize the biological activity of FGF2. Here, we demonstrate that ARPCA binds FGF8b and inhibits its capacity to form FGFR1-mediated ternary complexes with heparan sulphate proteoglycans. As a FGF8b antagonist, ARPCA inhibits FGFR1 activation and signalling in endothelial cells, hampering the angiogenic activity exerted in vitro and in vivo by FGF8b. Also, ARPCA suppresses the angiogenic and tumorigenic potential of prototypic androgen/FGF8b-dependent Shionogi 115 mammary carcinoma cells and of androgen/FGF8b/FGF2-dependent TRAMP-C2 prostate cancer cells. In conclusion, ARPCA represents a novel FGF8b antagonist with translational implications for the therapy of steroid hormone-regulated tumors.


Assuntos
Proteína C-Reativa/farmacologia , Fator 8 de Crescimento de Fibroblasto/metabolismo , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/metabolismo , Fragmentos de Peptídeos/farmacologia , Componente Amiloide P Sérico/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Fator 8 de Crescimento de Fibroblasto/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Modelos Moleculares , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neovascularização Fisiológica/efeitos dos fármacos
4.
Curr Opin Oncol ; 24(6): 666-71, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22759739

RESUMO

PURPOSE OF REVIEW: Outline current chemotherapy and targeted therapy treatments for intracranial meningiomas. RECENT FINDINGS: At present, there is no defined role for adjuvant chemotherapy for meningioma of any grade following initial diagnosis. In the subpopulation of patients with an unresectable meningioma and refractory to radiotherapy, hormonal chemotherapy-targeted therapy may be prescribed. Notwithstanding limited data, hydroxyurea, somatostatin analogues and interferon-α have been modestly successful in patients with recurrent meningiomas. Emerging targeted therapies, particularly angiogenic inhibitors, may prove useful in refractory meningiomas as recently demonstrated with sunitinib and novel somatostatin analogues. SUMMARY: A number of challenges are apparent with respect to the use of chemotherapy or targeted therapy for intracranial meningioma. First, there is very limited published literature that provides compelling evidence from which to determine appropriate therapy. Second, there is a paucity of clinical trials for patients with recurrent meningioma. Third, there remains a lack of agreement or standardization as to what constitutes a meaningful response to medical therapy recognizing these metrics differ between low-grade and high-grade recurrent meningioma. As a consequence, there remains a significant unmet need in neuro-oncology for defining the role of chemotherapy or targeted therapy in recurrent meningioma.


Assuntos
Neoplasias Meníngeas/tratamento farmacológico , Meningioma/tratamento farmacológico , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Humanos , Neoplasias Meníngeas/irrigação sanguínea , Neoplasias Meníngeas/metabolismo , Meningioma/irrigação sanguínea , Meningioma/metabolismo , Terapia de Alvo Molecular , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/metabolismo , Neovascularização Patológica/prevenção & controle , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Med Hypotheses ; 79(4): 474-7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22824092

RESUMO

Retroperitoneal sarcomas are rare and aggressive tumors with a negative prognosis as there is currently no satisfactory treatment for them. The only proven factor that can significantly increase the otherwise poor survival of sarcoma patients is the radically of resection. However, the completeness of resection is hindered by the hypervascularized nature of sarcomas and the frequent involvement of major blood vessels. In this context, we propose to operate on retroperitoneal sarcomas only with the use of extracorporeal circulation, applying vascular clamps above and below the tumor, even with short periods of hypothermic circulatory arrest in complex cases. This technique would allow the surgeon to achieve complete tumor resections, approach large blood vessels easier and perform sofisticated vascular reconstructions with no fear of hemorrhage which is fundamental to achieve a bloodless surgical field. Also, we speculate on the etiology of retroperitoneal sarcomas that appear mostly during the period of menopause/andropause. Although both estrogens and androgens have been incriminated in inducing various cancer types, including sarcomas, an endogenous estradiol cathabolyte has been shown to have anti-tumor effects. Considering that during menopause/andropause sex steroid levels actually decrease, our second working hypothesis is that the increasing follicle-stimulating hormone (FSH) and especially luteinizing hormone (LH) levels, together with the relative estrogen/androgen imbalance, may be the triggering cause. Also, a certain level of estrogens (Methoxyestradiol) may be essential in limiting tumor development and dedifferentiation. Given that extragonadal sarcomas appear to behave as endocrine tumors, a targeted hormonal therapy, together with controlled radical resections in complex cases of tumor vascular involvement, would certainly provide a strong link to both prevention and treatment of retroperitoneal sarcomas and even of cancer in general.


Assuntos
Neoplasias Retroperitoneais/cirurgia , Sarcoma/cirurgia , Carcinógenos/metabolismo , Estrogênios/metabolismo , Circulação Extracorpórea , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Hormônio Luteinizante/metabolismo , Masculino , Modelos Biológicos , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/etiologia , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/cirurgia , Neoplasias Retroperitoneais/irrigação sanguínea , Neoplasias Retroperitoneais/etiologia , Neoplasias Retroperitoneais/metabolismo , Sarcoma/irrigação sanguínea , Sarcoma/etiologia , Sarcoma/metabolismo
6.
Radiat Oncol ; 7: 75, 2012 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-22621752

RESUMO

BACKGROUND: Radiotherapy (RT) and androgen-deprivation therapy (ADT) are standard treatments for advanced prostate cancer (PC). Tumor vascularization is recognized as an important physiological feature likely to impact on both RT and ADT response, and this study therefore aimed to characterize the vascular responses to RT and ADT in experimental PC. METHODS: Using mice implanted with CWR22 PC xenografts, vascular responses to RT and ADT by castration were visualized in vivo by DCE MRI, before contrast-enhancement curves were analyzed both semi-quantitatively and by pharmacokinetic modeling. Extracted image parameters were correlated to the results from ex vivo quantitative fluorescent immunohistochemical analysis (qIHC) of tumor vascularization (9 F1), perfusion (Hoechst 33342), and hypoxia (pimonidazole), performed on tissue sections made from tumors excised directly after DCE MRI. RESULTS: Compared to untreated (Ctrl) tumors, an improved and highly functional vascularization was detected in androgen-deprived (AD) tumors, reflected by increases in DCE MRI parameters and by increased number of vessels (VN), vessel density (VD), and vessel area fraction (VF) from qIHC. Although total hypoxic fractions ( HF) did not change, estimated acute hypoxia scores (AHS)--the proportion of hypoxia staining within 50 µm from perfusion staining--were increased in AD tumors compared to in Ctrl tumors. Five to six months after ADT renewed castration-resistant (CR) tumor growth appeared with an even further enhanced tumor vascularization. Compared to the large vascular changes induced by ADT, RT induced minor vascular changes. Correlating DCE MRI and qIHC parameters unveiled the semi-quantitative parameters area under curve (AUC) from initial time-points to strongly correlate with VD and VF, whereas estimation of vessel size (VS) by DCE MRI required pharmacokinetic modeling. HF was not correlated to any DCE MRI parameter, however, AHS may be estimated after pharmacokinetic modeling. Interestingly, such modeling also detected tumor necrosis very strongly. CONCLUSIONS: DCE MRI reliably allows non-invasive assessment of tumors' vascular function. The findings of increased tumor vascularization after ADT encourage further studies into whether these changes are beneficial for combined RT, or if treatment with anti-angiogenic therapy may be a strategy to improve the therapeutic efficacy of ADT in advanced PC.


Assuntos
Vasos Sanguíneos/efeitos da radiação , Carcinoma/irrigação sanguínea , Carcinoma/radioterapia , Neoplasias Hormônio-Dependentes/radioterapia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/radioterapia , Androgênios/deficiência , Animais , Vasos Sanguíneos/fisiopatologia , Carcinoma/patologia , Carcinoma/cirurgia , Linhagem Celular Tumoral , Terapia Combinada , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/patologia , Neoplasias Experimentais/radioterapia , Neoplasias Experimentais/cirurgia , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/patologia , Neoplasias Hormônio-Dependentes/cirurgia , Neovascularização Patológica/radioterapia , Neovascularização Patológica/cirurgia , Orquiectomia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Endocr Relat Cancer ; 18(3): R41-51, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21367845

RESUMO

Sex hormones modulate proliferation, apoptosis, migration, metastasis and angiogenesis in cancer cells influencing tumourigenesis from the early hyperplastic growth till the end-stage metastasis. Although decades of studies have detailed these effects at the level of molecular pathways, where and when these actions are needed for the growth and progression of hormone-dependent neoplasia is poorly elucidated. Investigation of the hormone influences in carcinogenesis in the spatio-temporal dimension is expected to unravel critical steps in tumour progression and in the onset of resistance to hormone therapies. Non-invasive in vivo imaging represents a powerful tool to follow in time hormone signalling in the whole body during tumour development. This review summarizes the tools currently available to follow hormone action in living organisms.


Assuntos
Hormônios Esteroides Gonadais/fisiologia , Neoplasias Experimentais/fisiopatologia , Neoplasias Hormônio-Dependentes/fisiopatologia , Animais , Apoptose , Transformação Celular Neoplásica , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Reporter , Hormônios Esteroides Gonadais/efeitos adversos , Hormônios Esteroides Gonadais/biossíntese , Hormônios Esteroides Gonadais/toxicidade , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/metabolismo , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/metabolismo , Neovascularização Patológica/fisiopatologia , Receptores Androgênicos/genética , Receptores Androgênicos/fisiologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/genética , Receptores de Progesterona/fisiologia , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais
8.
Cancer Res ; 71(1): 51-60, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21097717

RESUMO

The proinflammatory cytokines IL-1α and IL-1ß promote tumor angiogenesis that might be counteracted by the IL-1 receptor antagonist (IL-1Ra), anakinra, a clinically approved agent. A diet with high amounts of phytoestrogens, such as flaxseed (Flax), genistein (GEN), and the mammalian lignan enterolactone (ENL), may affect breast cancer progression in a similar fashion as the antiestrogen tamoxifen. Both cancer cells and tumor stroma may be targets for cancer therapy. By using microdialysis in a model of human breast cancers in nude mice, we could perform species-specific analyses of released proteins in the microenvironment. We show that tumors treated with tamoxifen and fed Flax or ENL exhibited decreased in vivo release of IL-1ß derived from the murine stroma and decreased microvessel density whereas dietary GEN had no effects. Cancer cell-released IL-1Ra were approximately 5 times higher than stroma-derived IL-1Ra. Tamoxifen, Flax, and ENL increased IL-1Ra levels significantly whereas GEN did not. The tumor stroma contained macrophages, which expressed the estrogen receptor. In vitro, estradiol decreased IL-1Ra released from breast cancer cells and from cultured macrophages. IL-1Ra decreased endothelial cell proliferation significantly in vitro whereas breast cancer cell proliferation was unaffected in presence of estradiol. Finally, IL-1Ra therapy of tumor-bearing mice opposed estrogen-dependent breast cancer growth and decreased angiogenesis. We conclude that the release of IL-1s both by cancer cells and the stroma, where macrophages are a key component, may offer feasible targets for antiestrogen therapy and dietary interventions against breast cancer.


Assuntos
4-Butirolactona/análogos & derivados , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/irrigação sanguínea , Estrogênios/fisiologia , Linho , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Lignanas/farmacologia , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Células Estromais/efeitos dos fármacos , Tamoxifeno/farmacologia , 4-Butirolactona/farmacologia , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Hormônio-Dependentes/patologia , Células Estromais/metabolismo
9.
Neoplasia ; 12(10): 818-25, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20927320

RESUMO

Prostate cancer (PCa) patients receive androgen-deprivation therapy (ADT) to reduce tumor burden. However, complete eradication of PCa is unusual, and recurrent disease is evident within approximately 2 years in high-risk patients. Clinical evidence suggests that combining ADT with radiotherapy improves local control and disease-free survival in these patients compared with radiotherapy alone. We investigated whether vascularization of androgen-sensitive PCa xenografts changed after ADT and whether such therapy affected radiation response. CWR22 xenografts received combinations of ADT by castration (CWR22-cas) and 15 Gy of single-dose irradiation. At a shortest tumor diameter of 8 mm, vascularization was visualized by dynamic contrast-enhanced magnetic resonance imaging before radiation and 1 and 9 days after radiation. Voxel-wise quantitative modeling of contrast enhancement curves extracted the hemodynamic parameter K(trans), reflecting a combination of permeability, density, and blood flow. Tumor volumes and prostate-specific antigen (PSA) were monitored during the experiment. The results showed that K(trans) of CWR22-cas tumors 36±4 days after ADT was 47.1% higher than K(trans) of CWR22 tumors (P = .01). CWR22-cas tumors showed no significant changes in K(trans) after radiation, whereas K(trans) of CWR22 tumors at day 1 decreased compared with pretreatment values (P = .04) before a continuous increase from day 1 to day 9 followed (P = .01). Total PSA in blood correlated positively to tumor volume (r = 0.59, P < .01). In conclusion, androgen-exposed xenografts demonstrated radiation-induced reductions in vascularization and tumor volumes, whereas androgen-deprived xenografts showed increased vascularization and growth inhibition, but no significant additive effect of radiation.


Assuntos
Androgênios/metabolismo , Imageamento por Ressonância Magnética , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/terapia , Neovascularização Patológica/diagnóstico , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/terapia , Animais , Castração , Radioisótopos de Cobalto/uso terapêutico , Terapia Combinada , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Hormônio-Dependentes/radioterapia , Neoplasias Hormônio-Dependentes/cirurgia , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/radioterapia , Neoplasias da Próstata/cirurgia , Taxa de Sobrevida , Resultado do Tratamento , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 70(7): 2655-64, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20332242

RESUMO

"Reactive" or activated stroma characterizes many malignancies including breast cancers. Recently, we isolated a reactive mouse mammary gland stromal cell line called BJ3Z. These cells express alpha-smooth muscle actin and stromal cell-derived factor 1 (SDF-1) and are tumorigenic when injected into mice. Here we show that, in vivo, BJ3Z cells influence the angiogenesis and proliferation of xenografted estrogen receptor (ER)-positive MCF-7 human breast cancer cell-derived solid tumors. The growth-promoting effects of BJ3Z cells are equivalent to those of estradiol (E(2)). BJ3Z cells also increase the proliferation of normal mouse mammary luminal cells adjacent to tumors. In vitro, BJ3Z cells reorganize and increase the proliferation of cocultured malignant MCF-7 and normal human breast MCF10A cells grown as organoids in three-dimensional culture. The effects of BJ3Z cells on MCF-7 cells are equivalent to those of E(2). In contrast, BJ3Z cells do not alter the growth of highly aggressive ER-negative MDA-MB-231 human breast cancer cells. We show that BJ3Z cells secrete vascular endothelial growth factor (VEGF). The growth of MCF-7 organoids induced by BJ3Z can be inhibited by antagonists of VEGF and SDF-1. Conversely, recombinant VEGF stimulates the proliferation of MCF-7, but not MDA-MB-231, organoids. We conclude that, in addition to angiogenesis, VEGF released by activated stroma increases the growth of ER-positive malignant epithelial cells and of adjacent normal epithelium. Because activated stroma can substitute for E(2) and fosters hormone-independent growth of ER-positive tumors, we suggest that breast cancers exhibiting intrinsic hormone resistance may respond to antiangiogenic therapies.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Receptores de Estrogênio/biossíntese , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Quimiocina CXCL12/biossíntese , Células Epiteliais/patologia , Feminino , Humanos , Camundongos , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Células Estromais/metabolismo , Células Estromais/patologia
11.
Breast Cancer Res Treat ; 118(1): 33-43, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18855134

RESUMO

To investigate the therapeutic effect of methylselenocysteine (MSC) combined with tamoxifen in MCF-7 breast cancer xenograft and the underlying mechanisms. MCF-7 breast cancer xenograft was established in ovariectomized female athymic nude mice and treated with tamoxifen and/or MSC. Tumor size was measured twice a week. Immunohistochemistry and TUNEL assays were used to measure ERalpha expression, ERalpha target genes (progesterone receptor (PR) and cyclin D1 expression), Ki-67 index, apoptosis and microvessel density. Combined treatment with tamoxifen and MSC synergistically inhibited tumor growth compared to MSC alone and tamoxifen alone. MSC alone or MSC + tamoxifen significantly reduced ERalpha, PR and cyclin D1, Ki67 index and microvessel density while increasing apoptosis in tumor tissues. These findings demonstrate synergistic growth inhibition of ERalpha positive breast cancer xenografts by combination of tamoxifen with organic selenium compounds. Organic selenium may provide added benefit when combined with tamoxifen in adjuvant therapy or prevention.


Assuntos
Adenocarcinoma/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Cisteína/análogos & derivados , Estradiol , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Compostos Organosselênicos/farmacologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Ciclina D1/biossíntese , Ciclina D1/genética , Cisteína/administração & dosagem , Cisteína/farmacologia , Sinergismo Farmacológico , Estradiol/toxicidade , Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Compostos Organosselênicos/administração & dosagem , Distribuição Aleatória , Receptores de Progesterona/biossíntese , Receptores de Progesterona/genética , Selenocisteína/análogos & derivados , Organismos Livres de Patógenos Específicos , Tamoxifeno/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
12.
BJU Int ; 102(8): 1034-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18489523

RESUMO

OBJECTIVE: To investigate the relationship between microvessel density (MVD), blood vessel morphology and the expression of angiopoietin (Ang)-1, Ang-2, tyrosine kinase with immunoglobulin and epidermal growth factor homology domains (Tie)-2, and vascular endothelial growth factor (VEGF) in androgen-dependent (AD) and androgen-independent (AI) prostate cancer models, to gain insight into the regulation of angiogenesis at different stages of prostate cancer. MATERIALS AND METHODS: MVD and blood vessel morphology were evaluated by CD34 immunohistochemical staining. The mRNA and protein secretion of the Angs, Tie-2 and VEGF were measured by real-time polymerase chain reaction and enzyme-linked immunosorbent assays, respectively, in LNCaP (AD) and LNCaP-19, C4-2, C4-2B4 and PC-3 (AI) prostate cancer xenografts in mice. RESULTS: LNCaP, C4-2 and C4-2B4 xenografts had high expression of Ang-2 and VEGF, similar MVD and blood vessel morphology. However, the most angiogenic cell line LNCaP-19 expressed low levels of both factors and had different vessel morphology. PC-3 xenografts had a similar MVD to LNCaP, C4-2 and C4-2B4, but the Ang-2 and VEGF expression as well as the vessel morphology were similar to LNCaP-19. CONCLUSION: The differences in MVD, blood vessel morphology and the expression of Ang-2 and VEGF show that prostate cancer cells display angiogenic heterogeneity, which indicates different roles of these factors in the regulation of angiogenesis in different stages of prostate cancer.


Assuntos
Angiopoietina-2/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neovascularização Patológica/metabolismo , Neoplasias da Próstata/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/metabolismo , Androgênios/metabolismo , Angiopoietina-1/metabolismo , Animais , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Neovascularização Patológica/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Ann Oncol ; 19(4): 746-51, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18056648

RESUMO

BACKGROUND: The purpose of this trial was to evaluate the antitumor activity of sorafenib, a multikinase inhibitor of cell proliferation and angiogenesis, in patients with castration-resistant prostate cancer. PATIENTS AND METHODS: This was a multicenter, two-stage, phase II study. Sorafenib 400 mg was administered orally twice daily continuously. Primary end point was prostate-specific antigen (PSA) 'response' defined as a > or =50% decrease for > or =4 weeks. RESULTS: In all, 28 patients were enrolled. Eastern Cooperative Oncology Group performance status was zero or one in 19 and 9 patients. Two patients had no metastases, and 26 had bone and/or lymph node disease. A median of two cycles (range 1-8) was delivered. Adverse events were typical for sorafenib. The PSA response rate was 3.6% [95% confidence interval (CI) 0.1% to 18.3%] with response occurring in one patient (baseline = 10 000 and nadir = 1643 microg/l). No measurable disease responses occurred in eight patients. Time to PSA progression was 2.3 months (95% CI 1.8-6.4). Of 16 patients who discontinued sorafenib and then did not receive any immediate therapy, 10 had postdiscontinuation PSA declines of 7%-52%. CONCLUSIONS: Sorafenib has limited activity using current PSA criteria. The declines in PSA observed on treatment discontinuation indicate an effect on PSA production/secretion. Further study may be warranted but needs to consider the limitations of PSA as an indicator of progression and response.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Antineoplásicos/uso terapêutico , Benzenossulfonatos/uso terapêutico , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Administração Oral , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/administração & dosagem , Antineoplásicos/administração & dosagem , Benzenossulfonatos/administração & dosagem , Biomarcadores Tumorais/análise , Canadá , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Intervalo Livre de Doença , Esquema de Medicação , Resistencia a Medicamentos Antineoplásicos , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/química , Neoplasias Hormônio-Dependentes/imunologia , Neoplasias Hormônio-Dependentes/patologia , Neovascularização Patológica/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/química , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/administração & dosagem , Piridinas/administração & dosagem , Sorafenibe , Resultado do Tratamento
14.
Cancer Res ; 67(20): 9929-36, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17942925

RESUMO

Recent clinical trials indicate that synthetic progestins may stimulate progression of breast cancer in postmenopausal women, a result that is consistent with studies in chemically-induced breast cancer models in rodents. However, progestin-dependent progression of breast cancer tumor xenografts has not been shown. This study shows that xenografts obtained from BT-474 and T47-D human breast cancer cells without Matrigel in estrogen-supplemented nude mice begin to regress within days after tumor cell inoculation. However, their growth is resumed if animals are supplemented with progesterone. The antiprogestin RU-486 blocks progestin stimulation of growth, indicating involvement of progesterone receptors. Exposure of xenografts to medroxyprogesterone acetate, a synthetic progestin used in postmenopausal hormone replacement therapy and oral contraception, also stimulates growth of regressing xenograft tumors. Tumor progression is dependent on expression of vascular endothelial growth factor (VEGF); growth of progestin-dependent tumors is blocked by inhibiting synthesis of VEGF or VEGF activity using a monoclonal anti-VEGF antibody (2C3) or by treatment with PRIMA-1, a small-molecule compound that reactivates mutant p53 into a functional protein and blocks VEGF production. These results suggest a possible model system for screening potential therapeutic agents for their ability to prevent or inhibit progestin-dependent human breast tumors. Such a model could potentially be used to screen for safer antiprogestins, antiangiogenic agents, or for compounds that reactivate mutant p53 and prevent progestin-dependent progression of breast disease.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/patologia , Progestinas/antagonistas & inibidores , Progestinas/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Compostos Aza/farmacologia , Neoplasias da Mama/irrigação sanguínea , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Progressão da Doença , Feminino , Humanos , Acetato de Medroxiprogesterona/farmacologia , Camundongos , Mifepristona/farmacologia , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Progesterona/farmacologia , Congêneres da Progesterona/farmacologia , Receptores de Estrogênio/biossíntese , Receptores de Progesterona/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Neoplasia ; 9(7): 563-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17710159

RESUMO

Vessel size imaging was used to assess changes in the average vessel size of Shionogi tumors throughout the tumor growth cycle. Changes in R(2) and R(2)* relaxivities caused by the injection of a superparamagnetic contrast agent (ferumoxtran-10) were measured using a 2.35-T animal magnetic resonance imaging system, and average vessel size index (VSI) was calculated for each stage of tumor progression: growth, regression, and relapse. Statistical analysis using Spearman rank correlation test showed no dependence between vessel size and tumor volume at any stage of the tumor growth cycle. Paired Student's t test was used to assess the statistical significance of the differences in average vessel size for the three stages of the tumor growth cycle. The average VSI for regressing tumors (15.1 +/- 6.6 microm) was significantly lower than that for growing tumors (35.2 +/- 25.5 microm; P < .01). Relapsing tumors also had an average VSI (45.4 +/- 41.8 microm) higher than that of regressing tumors, although the difference was not statistically significant (P = .067). This study shows that VSI imaging is a viable method for the noninvasive monitoring of angiogenesis during the progression of a Shionogi tumor from androgen dependence to androgen independence.


Assuntos
Angiografia por Ressonância Magnética/métodos , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neovascularização Patológica/diagnóstico , Neoplasias da Próstata/irrigação sanguínea , Animais , Meios de Contraste/administração & dosagem , Dextranos , Modelos Animais de Doenças , Óxido Ferroso-Férrico , Ferro , Estudos Longitudinais , Nanopartículas de Magnetita , Masculino , Camundongos , Camundongos Endogâmicos , Transplante de Neoplasias , Neovascularização Patológica/patologia , Óxidos
16.
BioDrugs ; 21(4): 215-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17628119

RESUMO

Thomas Beatson's celebrated description in 1896 of bilateral oophorectomy as effective therapy for premenopausal breast cancer could be considered as the first demonstration of response of any cancer to a 'targeted therapy.' At that time, however, the understanding of the mechanism of the intervention was minimal. In recent years a host of new rationally designed, molecularly targeted cancer therapies have been introduced from both large pharmaceutic and small biotechnology companies, and the portfolio of new targeted treatments in the pipeline appears to be unending. The existence of this array of potential new therapies is the result of a prodigious effort in the researching and defining of the molecular components of the cancer phenotype, and the subsequent rational design of agents to target candidate pathways. Experience with endocrine therapy has shown that targeted therapies require the target to be not merely expressed in the cancer phenotype, but important in regulating growth of cancer cells. We may well look back at many of the current targeted therapy trials as unrealistically simplistic in failing to adequately and define the target phenotype. This approach risks rejecting highly active treatments for a small subgroup of a study population where minimal activity is present for the majority. The future for breast cancer therapy is promising, but it is important to be prepared for disappointment, as early success in animal models cannot guarantee a successful human therapy. Stunning results such as the adjuvant trastuzumab trials are likely to remain the exceptions rather that the rule, and most gains will be modest advances. A better understanding of the molecular biology of cancer may also aid in guiding the most appropriate use of existing therapies such as conventional chemotherapy. This knowledge will facilitate the rational selection of drug combinations and/or sequencing based on their mechanisms of action at a molecular level. The aim of this paper is to review the current state-of-the-art in novel targeted therapies for breast cancer based on an understanding of this disease at the molecular level, with particular reference to those agents entering the clinic.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/irrigação sanguínea , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/antagonistas & inibidores , Farnesiltranstransferase/antagonistas & inibidores , Feminino , Humanos , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neovascularização Patológica , Proteínas Tirosina Quinases/antagonistas & inibidores , Receptor ErbB-2/biossíntese , Transdução de Sinais , Tamoxifeno/uso terapêutico , Trastuzumab
17.
Cancer Res ; 67(4): 1636-44, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17308104

RESUMO

Tumor growth and metastasis depend on angiogenesis that requires the cofactor copper. Consistently, high levels of copper have been found in many types of human cancers, including prostate, breast, colon, and lung. Recent studies suggest that copper could be used as a novel selective target for cancer therapies. Clioquinol is capable of forming stable complexes with copper and currently used in clinics for treatment of Alzheimer's disease. Most recently, it has been reported that clioquinol possesses antitumor effects. However, the underlying molecular mechanism is unclear. We report here that after binding to copper, clioquinol can inhibit the proteasomal chymotrypsin-like activity, repress androgen receptor (AR) protein expression, and induce apoptotic cell death in human prostate cancer LNCaP and C4-2B cells. In addition, clioquinol alone exhibits similar effects in prostate cancer cell lines with elevated copper at concentrations similar to those found in patients. Addition of dihydrotestosterone did not affect clioquinol-mediated proteasome inhibition in both prostate cancer cell lines. However, dihydrotestosterone partially inhibited clioquinol-induced AR suppression and apoptosis only in androgen-dependent LNCaP cells. Animal studies show that clioquinol treatment significantly inhibits the growth of human prostate tumor C4-2B xenografts (by 66%), associated with in vivo proteasome inhibition, AR protein repression, angiogenesis suppression, and apoptosis induction. Our study provides strong evidence that clioquinol is able to target tumor proteasome in vivo in a copper-dependent manner, resulting in formation of an active AR inhibitor and apoptosis inducer that is responsible for its observed antiprostate tumor effect.


Assuntos
Antagonistas de Receptores de Andrógenos , Apoptose/efeitos dos fármacos , Clioquinol/farmacologia , Cobre/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Inibidores de Proteases/farmacologia , Animais , Linhagem Celular Tumoral , Clioquinol/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Inibidores de Proteases/metabolismo , Inibidores de Proteassoma , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Clin Transl Oncol ; 8(4): 231-41, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16648098

RESUMO

Molecular genetic evidence indicates that endometrial carcinoma likely develops as the result of a multistep process of oncogene activation and tumor suppressor gene inactivation. These molecular alterations appear to be specific for Type I (endometrioid) and Type II (non endometrioid) cancers. Type I cancers are characterized by mutation of PTEN, KRAS2, defects in DNA mismatch repair, as evidenced by the microsatellite instability phenotype, and a near diploid karyotype. Type II cancers often contain mutations of TP53 and Her-2/neu and are usually nondiploid. The clinical value of many of these molecular markers is now being tested and it may help to refine diagnosis and establish an accurate prognosis. Furthermore, some of these tumor biomarkers constitute the targets for emerging therapies. Transtuzumab against Her-2/neu and bevacizumab against VEGF overexpressing carcinomas are among the promising novel treatments. Additional translational research is needed to identify molecular and genetic alterations with potential for therapeutic interventions.


Assuntos
Adenocarcinoma/etiologia , Transformação Celular Neoplásica/genética , Neoplasias do Endométrio/etiologia , Neoplasias Hormônio-Dependentes/etiologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/genética , Adenocarcinoma/terapia , Antineoplásicos/uso terapêutico , Terapia Combinada , Reparo do DNA/genética , Neoplasias do Endométrio/irrigação sanguínea , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/terapia , Estrogênios , Feminino , Regulação Neoplásica da Expressão Gênica , Genes erbB-2 , Genes p53 , Instabilidade Genômica , Humanos , Histerectomia , Repetições de Microssatélites , Mutagênese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/fisiopatologia , Oncogenes , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/fisiologia , Ploidias , Progesterona , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/genética , Receptores de Progesterona/fisiologia
19.
Cancer Res ; 66(7): 3667-72, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585192

RESUMO

Direct proliferative effects of estrogen (E(2)) on estrogen receptor-positive tumors are well documented; however, the potential for E(2) to mediate effects selective for the host (i.e., angiogenesis, vascular permeability, or stromal effects), which influence tumor growth and/or metastasis, has received less attention. In this study, we examine the capacity for E(2) to promote tumor growth and/or metastasis independent of direct effects on tumor cells. In these studies, we distinguish host versus tumor compartment components of E(2) action in tumor growth and metastasis by analysis of E(2)-nonresponsive tumor cells implanted in ovariectomized (OVX) mice that contain s.c. implants of placebo (OVX) or E(2)-containing slow-release pellets (OVX + E(2)). We show that the D121 lung carcinoma cell line is E(2)-nonresponsive, and following s.c. implantation in OVX versus OVX + E(2) mice, E(2) action on the host compartment leads to an increase in spontaneous metastasis but not primary tumor growth or neovascularization. Similarly, experimental lung metastasis of E(2)-nonresponsive 4T1 mammary carcinoma cells also leads to increased tumor burden in the lungs of OVX + E(2) mice. These results suggest that the E(2) status of the host compartment influences late steps in tumor cell metastasis that can provide important insights into the role of E(2) in the tumor versus host compartments.


Assuntos
Neoplasias da Mama/patologia , Estradiol/toxicidade , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Neoplasias Hormônio-Dependentes/secundário , Animais , Neoplasias da Mama/irrigação sanguínea , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/biossíntese , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/patologia , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neoplasias Hormônio-Dependentes/induzido quimicamente , Neoplasias Hormônio-Dependentes/patologia , Neovascularização Patológica/patologia
20.
J Steroid Biochem Mol Biol ; 93(2-5): 161-5, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15860258

RESUMO

In general, tumors induce angiogenic factors specific to them, which leads to angiogenesis with advancement. However, angiogenesis in uterine endometrial cancers is complicated because hormone dependency in growth also modifies the angiogenic potential. Therefore, anti-angiogenic therapy for tumor dormancy in uterine endometrial cancers must be thoroughly considered. The upstream of vascular endothelial growth factor (VEGF) gene conserves estrogen-responsive elements. Progesterone primed with estrogen induces thymidine phosphorylase (TP) in uterine endometrium. Sex steroid-dependent VEGF and TP are highly expressed in cases of early stage and well-differentiated uterine endometrial cancers, and basic fibroblast growth factor (bFGF) in cases of advanced and poorly differentiated uterine endometrial cancers. A transcriptional factor for angiogenesis, ETS-1, is linked to VEGF in well-differentiated uterine endometrial cancers, and to bFGF in poorly differentiated uterine endometrial cancers. Therefore, even if dedifferentiation and angiogenic switching occur due to advancement and long-term hormone therapy, the inhibition of ETS-1 along with main angiogenic factors might be an effective strategy to suppress uterine endometrial cancers as a novel anti-angiogenic therapy.


Assuntos
Neoplasias do Endométrio/irrigação sanguínea , Hormônios Esteroides Gonadais/metabolismo , Neoplasias Hormônio-Dependentes/irrigação sanguínea , Neovascularização Patológica/etiologia , Inibidores da Angiogênese/uso terapêutico , Linhagem Celular Tumoral , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Expressão Gênica , Humanos , Modelos Biológicos , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Proteína Proto-Oncogênica c-ets-1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Timidina Fosforilase/biossíntese , Timidina Fosforilase/genética , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA