Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Radiol Clin North Am ; 58(2): 401-412, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32044014

RESUMO

Gynecologic cancers impact women of all ages. Some women may wish to preserve their capacity for future childbearing. With appropriate patient selection, acceptable oncologic outcomes may be achieved with preservation of fertility. Determination of eligibility for fertility preservation is guided by patient factors, tumor histology, and preoperative local staging with pelvic MR imaging. The aim of this article is to educate radiologists on the current guidelines for fertility-sparing techniques in women with early stage cervical, endometrial, and ovarian malignancies.


Assuntos
Preservação da Fertilidade/métodos , Neoplasias dos Genitais Femininos/diagnóstico por imagem , Neoplasias dos Genitais Femininos/terapia , Imageamento por Ressonância Magnética Multiparamétrica/métodos , Adulto , Feminino , Genitália Feminina/diagnóstico por imagem , Humanos
2.
Mol Cancer Ther ; 18(1): 162-172, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30305341

RESUMO

For mucinous ovarian cancer (MOC), standard platinum-based therapy is largely ineffective. We sought to identify possible mechanisms of oxaliplatin resistance of MOC and develop strategies to overcome this resistance. A kinome-based siRNA library screen was carried out using human MOC cells to identify novel targets to enhance the efficacy of chemotherapy. In vitro and in vivo validations of antitumor effects were performed using mouse MOC models. Specifically, the role of PRKRA/PACT in oxaliplatin resistance was interrogated. We focused on PRKRA, a known activator of PKR kinase, and its encoded protein PACT because it was one of the five most significantly downregulated genes in the siRNA screen. In orthotopic mouse models of MOC, we observed a significant antitumor effect of PRKRA siRNA plus oxaliplatin. In addition, expression of miR-515-3p was regulated by PACT-Dicer interaction, and miR-515-3p increased the sensitivity of MOC to oxaliplatin. Mechanistically, miR-515-3p regulated chemosensitivity, in part, by targeting AXL. The PRKRA/PACT axis represents an important therapeutic target in MOC to enhance sensitivity to oxaliplatin.


Assuntos
Adenocarcinoma Mucinoso/patologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Ovarianas/patologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Regulação para Cima , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , RNA Helicases DEAD-box/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , MicroRNAs/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Oxaliplatina , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/farmacologia , Receptores Proteína Tirosina Quinases/genética , Ribonuclease III/metabolismo , Receptor Tirosina Quinase Axl
3.
Gynecol Oncol ; 151(3): 428-432, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30366647

RESUMO

OBJECTIVES: To determine the concordance between the laparoscopic scoring assessment and extent of disease identified at primary tumor reductive surgery (TRS) in patients with advanced ovarian cancer. METHODS: From April 2013 to June 2017, we prospectively triaged patients with stage IIA to IVB ovarian cancer to laparoscopic scoring assessment. A validated predictive index value (PIV) score (range: 0-14) was assigned. Patients with PIV scores <8 were offered primary surgery and those with score ≥8 received NACT. Patients who underwent primary TRS had a second PIV score based on laparotomy findings. Concordance percentages were calculated between the two scores. Positive predictive value (PPV) was calculated to reflect the performance of the laparoscopic PIV score to predict R0 (complete gross resection) at TRS. RESULTS: 226 patients underwent laparoscopic scoring assessment, of which 139 (61.5%) had a PIV score <8 and 81 (35.8%) had a PIV score ≥8. 6 patients (2.7%) were unscoreable. There was 96% overall concordance between PIV scores at laparoscopy and primary TRS. Concordance scores by location were: bowel infiltration 74.7%, mesenteric disease 84.6%, liver surface involvement 86.5%, omental disease 89.7%, diaphragm disease 92.9%, stomach infiltration 94.7%, peritoneal carcinomatosis 94.8%. A laparoscopic PIV score of <8 had a PPV of 85.4% at predicting R0 at primary TRS. CONCLUSIONS: Laparoscopic assessment of tumor burden is a feasible tool to predict R0 cytoreduction in patients with advanced ovarian cancer. Concordance between PIV scores at laparoscopy and primary TRS varied by anatomic location, with the lowest concordance seen in predicting bowel infiltration.


Assuntos
Procedimentos Cirúrgicos de Citorredução/métodos , Laparoscopia/métodos , Neoplasias Ovarianas/cirurgia , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Estudos Prospectivos , Adulto Jovem
4.
Mol Cancer Ther ; 17(2): 464-473, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29237804

RESUMO

Although progesterone receptor (PR)-targeted therapies are modestly active in patients with uterine cancer, their underlying molecular mechanisms are not well understood. The clinical use of such therapies is limited because of the lack of biomarkers that predict response to PR agonists (progestins) or PR antagonists (onapristone). Thus, understanding the underlying molecular mechanisms of action will provide an advance in developing novel combination therapies for cancer patients. Nuclear translocation of PR has been reported to be ligand-dependent or -independent. Here, we identified that onapristone, a PR antagonist, inhibited nuclear translocation of ligand-dependent or -independent (EGF) phospho-PR (S294), whereas trametinib inhibited nuclear translocation of EGF-induced phospho-PR (S294). Using orthotopic mouse models of uterine cancer, we demonstrated that the combination of onapristone and trametinib results in superior antitumor effects in uterine cancer models compared with either monotherapy. These synergistic effects are, in part, mediated through inhibiting the nuclear translocation of EGF-induced PR phosphorylation in uterine cancer cells. Targeting MAPK-dependent PR activation with onapristone and trametinib significantly inhibited tumor growth in preclinical uterine cancer models and is worthy of further clinical investigation. Mol Cancer Ther; 17(2); 464-73. ©2017 AACR.


Assuntos
Antineoplásicos/uso terapêutico , Gonanos/uso terapêutico , Receptores de Progesterona/antagonistas & inibidores , Neoplasias Uterinas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Gonanos/farmacologia , Humanos , Camundongos , Camundongos Nus , Neoplasias Uterinas/genética , Neoplasias Uterinas/patologia
5.
Oncotarget ; 8(57): 96496-96505, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29228548

RESUMO

Anti-angiogenesis therapy has shown clinical benefit in patients with high-grade serous ovarian cancer (HGSC), but adaptive resistance rapidly emerges. Thus, approaches to overcome such resistance are needed. We developed the setting of adaptive resistance to anti-VEGF therapy, and performed a series of in vivo experiments in both immune competent and nude mouse models. Given the pro-angiogenic properties of tumor-associated macrophages (TAMs) and the dominant role of CSF1R in macrophage function, we added CSF1R inhibitors following emergence of adaptive resistance to anti-VEGF antibody. Mice treated with a CSF1R inhibitor (AC708) after anti-VEGF antibody resistance had little to no measurable tumor burden upon completion of the experiment while those that did not receive a CSF1R inhibitor still had abundant tumor. To mimic clinically used regimens, mice were also treated with anti-VEGF antibody and paclitaxel until resistance emerged, and then a CSF1R inhibitor was added. The addition of a CSF1R inhibitor restored response to anti-angiogenesis therapy, resulting in 83% lower tumor burden compared to treatment with anti-VEGF antibody and paclitaxel alone. Collectively, our data demonstrate that the addition of a CSF1R inhibitor to anti-VEGF therapy and taxane chemotherapy results in robust anti-tumor effects.

6.
Nat Commun ; 8(1): 310, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28827520

RESUMO

Thrombocytosis is present in more than 30% of patients with solid malignancies and correlates with worsened patient survival. Tumor cell interaction with various cellular components of the tumor microenvironment including platelets is crucial for tumor growth and metastasis. Although it is known that platelets can infiltrate into tumor tissue, secrete pro-angiogenic and pro-tumorigenic factors and thereby increase tumor growth, the precise molecular interactions between platelets and metastatic cancer cells are not well understood. Here we demonstrate that platelets induce resistance to anoikis in vitro and are critical for metastasis in vivo. We further show that platelets activate RhoA-MYPT1-PP1-mediated YAP1 dephosphorylation and promote its nuclear translocation which induces a pro-survival gene expression signature and inhibits apoptosis. Reduction of YAP1 in cancer cells in vivo protects against thrombocytosis-induced increase in metastasis. Collectively, our results indicate that cancer cells depend on platelets to avoid anoikis and succeed in the metastatic process.Platelets have been associated with increased tumor growth and metastasis but the mechanistic details of this interaction are still unclear. Here the authors show that platelets improve anoikis resistance of cancer cells and increase metastasis by activating Yap through a RhoA/MYPT-PP1 pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Anoikis , Plaquetas/metabolismo , Neoplasias Ovarianas/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Plaquetas/citologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos Endogâmicos C57BL , Camundongos Nus , Metástase Neoplásica , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fosfoproteínas/genética , Interferência de RNA , Fatores de Transcrição , Transplante Heterólogo , Proteínas de Sinalização YAP
7.
J Natl Cancer Inst ; 109(7)2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28376174

RESUMO

Background: The PI3K/AKT/P70S6K pathway is an attractive therapeutic target in ovarian and uterine malignancies because of its high rate of deregulation and key roles in tumor growth. Here, we examined the biological effects of MSC2363318A, which is a novel inhibitor of AKT1, AKT3, and P70S6K. Methods: Orthotopic murine models of ovarian and uterine cancer were utilized to study the effect of MSC2363318A on survival and regression. For each cell line, 10 mice were treated in each of the experimental arms tested. Moreover, in vitro experiments in 21 cell lines (MTT, immunoblot analysis, plasmid transfection, reverse phase protein array [RPPA]) were carried out to characterize underlying mechanisms and potential biomarkers of response. All statistical tests were two-sided. Results: MSC2363318A decreased tumor growth and metastases in multiple murine orthotopic models of ovarian (SKOV3ip1, HeyA8, and Igrov1) and uterine (Hec1a) cancer by reducing proliferation and angiogenesis and increasing cell death. Statistically significant prolonged overall survival was achieved with combination MSC2363318A and paclitaxel in the SKUT2 (endometrioid) uterine cancer mouse model ( P < .001). Mice treated with combination MSC2363318A and paclitaxel had the longest overall survival (mean = 104.2 days, 95% confidence interval [CI] = 97.0 to 111.4) compared with those treated with vehicle (mean = 61.9 days, 95% CI = 46.3 to 77.5), MSC2363318A alone (mean = 89.7 days, 95% CI = 83.0 to 96.4), and paclitaxel alone (mean = 73.6 days, 95% CI = 53.4 to 93.8). Regression and stabilization of established tumors in the Ishikawa (endometrioid) uterine cancer model was observed in mice treated with combination MSC2363318A and paclitaxel. Synergy between MSC2363318A and paclitaxel was observed in vitro in cell lines that had an IC50 of 5 µM or greater. RPPA results identified YAP1 as a candidate marker to predict cell lines that were most sensitive to MSC2363318A (R = 0.54, P = .02). After establishment of a murine ovarian cancer model of adaptive anti-angiogenic resistance (SKOV3ip1-luciferase), we demonstrate that resensitization to bevacizumab occurs with the addition of MSC2363318A, resulting in improved overall survival ( P = .01) using the Kaplan-Meier method. Mice treated with bevacizumab induction followed by MSC2363318A had the longest overall survival (mean = 66.0 days, 95% CI = 53.9 to 78.1) compared with mice treated with control (mean = 42.0 days, 95% CI = 31.4 to 52.6) and bevacizumab-sensitive mice (mean = 47.2 days; 95% CI = 37.5 to 56.9). Conclusions: MSC2363318A has therapeutic efficacy in multiple preclinical models of ovarian and uterine cancer. These findings support clinical development of a dual AKT/P70S6K inhibitor.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias Ovarianas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Neoplasias Uterinas/metabolismo , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Bevacizumab/administração & dosagem , Bevacizumab/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Concentração Inibidora 50 , Estimativa de Kaplan-Meier , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Fatores de Transcrição , Carga Tumoral/efeitos dos fármacos , Neoplasias Uterinas/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas de Sinalização YAP
8.
Mol Cancer Ther ; 15(12): 2894-2904, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27638860

RESUMO

To determine the efficacy of a novel and safer (for gastrointestinal tract) aspirin (aspirin-PC) in preclinical models of ovarian cancer, in vitro dose-response studies were performed to compare the growth-inhibitory effect of aspirin-PC versus aspirin on three human (A2780, SKOV3ip1, and HeyA8) and a mouse (ID8) ovarian cancer cell line over an 8-day culture period. In the in vivo studies, the aspirin test drugs were studied alone and in the presence of a VEGF-A inhibitor (bevacizumab or B20), due to an emerging role for platelets in tumor growth following antiangiogenic therapy, and we examined their underlying mechanisms. Aspirin-PC was more potent (vs. aspirin) in blocking the growth of both human and mouse ovarian cancer cells in monolayer culture. Using in vivo model systems of ovarian cancer, we found that aspirin-PC significantly reduced ovarian cancer growth by 50% to 90% (depending on the ovarian cell line). The efficacy was further enhanced in combination with Bevacizumab or B20. The growth-inhibitory effect on ovarian tumor mass and number of tumor nodules was evident, but less pronounced for aspirin and the VEGF inhibitors alone. There was no detectable gastrointestinal toxicity. Both aspirin and aspirin-PC also inhibited cell proliferation, angiogenesis, and increased apoptosis of ovarian cancer cells. In conclusion, PC-associated aspirin markedly inhibits the growth of ovarian cancer cells, which exceeds that of the parent drug, in both cell culture and in mouse model systems. We also found that both aspirin-PC and aspirin have robust antineoplastic action in the presence of VEGF-blocking drugs. Mol Cancer Ther; 15(12); 2894-904. ©2016 AACR.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Aspirina/farmacologia , Neovascularização Patológica , Neoplasias Ovarianas/patologia , Fosfatidilcolinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Tromboxanos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Clin Invest ; 126(5): 1885-96, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27064283

RESUMO

Recent studies in patients with ovarian cancer suggest that tumor growth may be accelerated following cessation of antiangiogenesis therapy; however, the underlying mechanisms are not well understood. In this study, we aimed to compare the effects of therapy withdrawal to those of continuous treatment with various antiangiogenic agents. Cessation of therapy with pazopanib, bevacizumab, and the human and murine anti-VEGF antibody B20 was associated with substantial tumor growth in mouse models of ovarian cancer. Increased tumor growth was accompanied by tumor hypoxia, increased tumor angiogenesis, and vascular leakage. Moreover, we found hypoxia-induced ADP production and platelet infiltration into tumors after withdrawal of antiangiogenic therapy, and lowering platelet counts markedly inhibited tumor rebound after withdrawal of antiangiogenic therapy. Focal adhesion kinase (FAK) in platelets regulated their migration into the tumor microenvironment, and FAK-deficient platelets completely prevented the rebound tumor growth. Additionally, combined therapy with a FAK inhibitor and the antiangiogenic agents pazopanib and bevacizumab reduced tumor growth and inhibited negative effects following withdrawal of antiangiogenic therapy. In summary, these results suggest that FAK may be a unique target in situations in which antiangiogenic agents are withdrawn, and dual targeting of FAK and VEGF could have therapeutic implications for ovarian cancer management.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Quinase 1 de Adesão Focal/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Difosfato de Adenosina/metabolismo , Animais , Anticorpos Antineoplásicos/farmacologia , Bevacizumab/farmacologia , Plaquetas/patologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Linhagem Celular Tumoral , Feminino , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/genética , Humanos , Indazóis , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Neovascularização Patológica/enzimologia , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Pirimidinas/farmacologia , Sulfonamidas/farmacologia , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Eur J Cancer ; 56: 131-143, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26849037

RESUMO

The study of cancer initiation, growth, and metastasis has traditionally been focused on cancer cells, and the view that they proliferate due to uncontrolled growth signalling owing to genetic derangements. However, uncontrolled growth in tumours cannot be explained solely by aberrations in cancer cells themselves. To fully understand the biological behaviour of tumours, it is essential to understand the microenvironment in which cancer cells exist, and how they manipulate the surrounding stroma to promote the malignant phenotype. Ovarian cancer is the leading cause of death from gynaecologic cancer worldwide. The majority of patients will have objective responses to standard tumour debulking surgery and platinum-taxane doublet chemotherapy, but most will experience disease recurrence and chemotherapy resistance. As such, a great deal of effort has been put forth to develop therapies that target the tumour microenvironment in ovarian cancer. Herein, we review the key components of the tumour microenvironment as they pertain to this disease, outline targeting opportunities and supporting evidence thus far, and discuss resistance to therapy.


Assuntos
Antineoplásicos/uso terapêutico , Terapia de Alvo Molecular , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Inibidores da Angiogênese/uso terapêutico , Animais , Carcinoma Epitelial do Ovário , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Neoplasias Epiteliais e Glandulares/irrigação sanguínea , Neoplasias Epiteliais e Glandulares/imunologia , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neovascularização Patológica , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos
11.
Clin Cancer Res ; 22(7): 1713-24, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26581245

RESUMO

PURPOSE: Chronic adrenergic activation has been shown to associate with adverse clinical outcomes in cancer patients, but the underlying mechanisms are not well understood. The focus of the current study was to determine the functional and biologic effects of adrenergic pathways on response to chemotherapy in the context of ovarian cancer. EXPERIMENTAL DESIGN: Increased DUSP1 production by sympathetic nervous system mediators (e.g., norepinephrine) was analyzed by real-time quantitative RT-PCR and by Western blotting. In vitro chemotherapy-induced cell apoptosis was examined by flow cytometry. For in vivo therapy, a well-characterized model of chronic stress was used. RESULTS: Catecholamines significantly inhibited paclitaxel- and cisplatin-induced apoptosis in ovarian cancer cells. Genomic analyses of cells treated with norepinephrine identified DUSP1 as a potential mediator. DUSP1 overexpression resulted in reduced paclitaxel-induced apoptosis in ovarian cancer cells compared with control; conversely, DUSP1 gene silencing resulted in increased apoptosis compared with control cells. DUSP1 gene silencing in vivo significantly enhanced response to paclitaxel and increased apoptosis. In vitro analyses indicated that norepinephrine-induced DUSP1 gene expression was mediated through ADRB2 activation of cAMP-PLC-PKC-CREB signaling, which inhibits JNK-mediated phosphorylation of c-Jun and protects ovarian cancer cells from apoptosis. Moreover, analysis of The Cancer Genome Atlas data showed that increased DUSP1 expression was associated with decreased overall (P= 0.049) and progression-free (P= 0.0005) survival. CONCLUSIONS: These findings provide a new understanding of the mechanisms by which adrenergic pathways can impair response to chemotherapy and have implications for cancer management.


Assuntos
Adrenérgicos/farmacologia , Antineoplásicos/farmacologia , Fosfatase 1 de Especificidade Dupla/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Catecolaminas/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fosfatase 1 de Especificidade Dupla/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Modelos Biológicos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico , Transcrição Gênica , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Cancer Ther ; 14(12): 2677-86, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26516159

RESUMO

There is growing recognition of the important role of metronomic chemotherapy in cancer treatment. On the basis of their unique antiangiogenic effects, we tested the efficacy of nab-paclitaxel, which stimulates thrombospondin-1, and topotecan, which inhibits hypoxia-inducible factor 1-α, at metronomic dosing for the treatment of ovarian carcinoma. In vitro and in vivo SKOV3ip1, HeyA8, and HeyA8-MDR (taxane-resistant) orthotopic models were used to examine the effects of metronomic nab-paclitaxel and metronomic topotecan. We examined cell proliferation (Ki-67), apoptosis (cleaved caspase-3), and angiogenesis (microvessel density, MVD) in tumors obtained at necropsy. In vivo therapy experiments demonstrated treatment with metronomic nab-paclitaxel alone and in combination with metronomic topotecan resulted in significant reductions in tumor weight (62% in the SKOV3ip1 model, P < 0.01 and 96% in the HeyA8 model, P < 0.03) compared with vehicle (P < 0.01). In the HeyA8-MDR model, metronomic monotherapy with either cytotoxic agent had modest effects on tumor growth, but combination therapy decreased tumor burden by 61% compared with vehicle (P < 0.03). The greatest reduction in MVD (P < 0.05) and proliferation was seen in combination metronomic therapy groups. Combination metronomic therapy resulted in prolonged overall survival in vivo compared with other groups (P < 0.001). Tube formation was significantly inhibited in RF-24 endothelial cells exposed to media conditioned with metronomic nab-paclitaxel alone and media conditioned with combination metronomic nab-paclitaxel and metronomic topotecan. The combination of metronomic nab-paclitaxel and metronomic topotecan offers a novel, highly effective therapeutic approach for ovarian carcinoma that merits further clinical development.


Assuntos
Albuminas/administração & dosagem , Neovascularização Patológica/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/administração & dosagem , Topotecan/administração & dosagem , Administração Metronômica , Inibidores da Angiogênese/administração & dosagem , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Camundongos , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Cell ; 28(5): 610-622, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26481148

RESUMO

While recombinant human erythropoietin (rhEpo) has been widely used to treat anemia in cancer patients, concerns about its adverse effects on patient survival have emerged. A lack of correlation between expression of the canonical EpoR and rhEpo's effects on cancer cells prompted us to consider the existence of an alternative Epo receptor. Here, we identified EphB4 as an Epo receptor that triggers downstream signaling via STAT3 and promotes rhEpo-induced tumor growth and progression. In human ovarian and breast cancer samples, expression of EphB4 rather than the canonical EpoR correlated with decreased disease-specific survival in rhEpo-treated patients. These results identify EphB4 as a critical mediator of erythropoietin-induced tumor progression and further provide clinically significant dimension to the biology of erythropoietin.


Assuntos
Neoplasias da Mama/genética , Eritropoetina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Ovarianas/genética , Receptor EphB4/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Progressão da Doença , Eritropoetina/genética , Feminino , Humanos , Estimativa de Kaplan-Meier , Células MCF-7 , Camundongos Endogâmicos C57BL , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ligação Proteica/efeitos dos fármacos , Receptor EphB4/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Adulto Jovem
14.
Eur J Cancer ; 51(14): 1978-88, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26238017

RESUMO

BACKGROUND: We compared survival outcomes and risk of venous thromboembolism (VTE) among patients with advanced and early-stage ovarian clear cell carcinoma (OCCC) and serous ovarian carcinoma (SOC), as well as potential links with interleukin-6 (IL-6) levels. METHODS: A multicenter case-control study was conducted in 370 patients with OCCC and 938 with SOC. In a subset of 200 cases, pretreatment plasma IL-6 levels were examined. FINDINGS: Patients with advanced OCCC had the highest 2-year cumulative VTE rates (advanced OCCC 43.1%, advanced SOC 16.2%, early-stage OCCC 11.9% and early-stage SOC 6.4%, P<0.0001) and the highest median levels of IL-6 (advanced OCCC 17.8 pg/mL, advanced SOC 9.0 pg/mL, early-stage OCCC 4.2 pg/mL and early-stage SOC 5.0 pg/mL, P=0.006). Advanced OCCC (hazard ratio [HR] 3.38, P<0.0001), thrombocytosis (HR 1.42, P=0.032) and elevated IL-6 (HR 8.90, P=0.046) were independent predictors of VTE. In multivariate analysis, patients with advanced OCCC had significantly poorer 5-year progression-free and overall survival rates than those with advanced SOC (P<0.01), and thrombocytosis was an independent predictor of decreased survival outcomes (P<0.01). Elevated IL-6 levels led to poorer 2-year progression-free survival rates in patients with OCCC (50% versus 87.5%, HR 4.89, P=0.016) than in those with SOC (24.9% versus 40.8%, HR 1.40, P=0.07). INTERPRETATION: Advanced OCCC is associated with an increased incidence of VTE and decreased survival outcomes, which has major implications for clinical management of OCCC.


Assuntos
Biomarcadores Tumorais/sangue , Carcinoma/complicações , Interleucina-6/sangue , Neoplasias Císticas, Mucinosas e Serosas/complicações , Neoplasias Ovarianas/complicações , Tromboembolia Venosa/etiologia , Adulto , Idoso , Carcinoma/sangue , Carcinoma/mortalidade , Carcinoma/patologia , Progressão da Doença , Intervalo Livre de Doença , Feminino , Humanos , Incidência , Japão , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Análise Multivariada , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Neoplasias Císticas, Mucinosas e Serosas/sangue , Neoplasias Císticas, Mucinosas e Serosas/mortalidade , Neoplasias Císticas, Mucinosas e Serosas/patologia , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Estados Unidos , Regulação para Cima , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/mortalidade
15.
Mol Cancer Ther ; 14(6): 1466-1475, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25833835

RESUMO

PTEN is known to be frequently mutated in uterine cancer and also dephosphorylates FAK. Here, we examined the impact of PTEN alterations on the response to treatment with a FAK inhibitor (GSK2256098). In vitro and in vivo therapeutic experiments were carried out using PTEN-mutated and PTEN-wild-type models of uterine cancer alone and in combination with chemotherapy. Treatment with GSK2256098 resulted in greater inhibition of pFAK(Y397) in PTEN-mutated (Ishikawa) than in PTEN-wild-type (Hec1A) cells. Ishikawa cells were more sensitive to GSK2256098 than the treated Hec1A cells. Ishikawa cells were transfected with a wild-type PTEN construct and pFAK(Y397) expression was unchanged after treatment with GSK2256098. Decreased cell viability and enhanced sensitivity to chemotherapy (paclitaxel and topotecan) in combination with GSK2256098 was observed in Ishikawa cells as compared with Hec1a cells. In the Ishikawa orthoptopic murine model, treatment with GSK2256098 resulted in lower tumor weights and fewer metastases than mice inoculated with Hec1A cells. Tumors treated with GSK2256098 had lower microvessel density (CD31), less cellular proliferation (Ki67), and higher apoptosis (TUNEL) rates in the Ishikawa model when compared with the Hec1a model. From a large cohort of evaluable patients, increased FAK and pFAK(Y397) expression levels were significantly related to poor overall survival. Moreover, PTEN levels were inversely related to pFAK(Y397) expression. These preclinical data demonstrate that PTEN-mutated uterine cancer responds better to FAK inhibition than does PTEN wild-type cancer. Therefore, PTEN could be a biomarker for predicting response to FAK-targeted therapy during clinical development.


Assuntos
Aminopiridinas/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Ácidos Hidroxâmicos/farmacologia , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias Uterinas/tratamento farmacológico , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Camundongos Nus , Mutação , Neovascularização Patológica/metabolismo , Neovascularização Patológica/prevenção & controle , PTEN Fosfo-Hidrolase/genética , Fosforilação/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Valor Preditivo dos Testes , Prognóstico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Uterinas/irrigação sanguínea , Neoplasias Uterinas/genética , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Oncotarget ; 6(6): 4266-73, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25738355

RESUMO

Increased adrenergic signaling facilitates tumor progression, but the underlying mechanisms remain poorly understood. We examined factors responsible for stress-mediated effects on monocyte/macrophage recruitment into the tumor microenvironment, and the resultant effects on tumor growth. In vitro, MCP1 was significantly increased after catecholamine exposure, which was mediated by cAMP and PKA. Tumor samples from mice subjected to daily restraint stress had elevated MCP1 gene and protein levels, increased CD14+ cells, and increased infiltration of CD68+ cells. hMCP1 siRNA-DOPC nanoparticles significantly abrogated daily restraint stress-induced tumor growth and inhibited infiltration of CD68+ and F4/80+ cells. In ovarian cancer patients, elevated peripheral blood monocytes and tumoral macrophages were associated with worse overall survival. Collectively, we demonstrate that increased adrenergic signaling is associated with macrophage infiltration and mediated by tumor cell-derived MCP1 production.


Assuntos
Carcinoma/patologia , Quimiocina CCL2/metabolismo , Macrófagos/metabolismo , Neoplasias Ovarianas/patologia , Estresse Psicológico/metabolismo , Animais , Carcinoma/metabolismo , Carcinoma/mortalidade , Quimiotaxia de Leucócito/fisiologia , Ensaio de Imunoadsorção Enzimática , Epinefrina/metabolismo , Feminino , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Norepinefrina/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Reação em Cadeia da Polimerase em Tempo Real
17.
Contraception ; 86(3): 281-4, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22364817

RESUMO

BACKGROUND: Intrauterine devices are the most used long-acting reversible contraceptive method worldwide. Under normal circumstances, removal of an intrauterine contraceptive (IUC) is an uncomplicated procedure requiring gentle traction on the string. STUDY DESIGN: We report three cases of nonvisible IUC strings where, following use of vaginal misoprostol, the IUC strings were visualized and the IUCs were removed intact with gentle traction. CONCLUSIONS: The uterotonic and uterocontractile effects following vaginal misoprostol facilitated removal in three cases of nonvisible IUC strings. We suggest that clinicians consider including vaginal misoprostol alone or prior to planned repeat office or procedure-clinic interventions for nonvisible IUC strings.


Assuntos
Remoção de Dispositivo/métodos , Dispositivos Intrauterinos , Misoprostol/uso terapêutico , Ocitócicos/uso terapêutico , Administração Intravaginal , Adulto , Feminino , Humanos
18.
Am J Respir Cell Mol Biol ; 34(5): 552-60, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16415250

RESUMO

The doxycycline-inducible, gene regulatory system allows tight control of transgene expression for the study of organ development and disease pathogenesis. Multiple recent reports have employed this model to investigate various lung diseases including emphysema. For our study, we used this transgenic system to test whether prolonged, lung-specific, overexpression of the serine protease urokinase plasminogen activator (uPA) would result in alveolar wall destruction. Double transgenic mice were generated that possessed: (1) the rat Clara cell secretory protein promoter controlling the reverse tetracycline transactivator gene (CCSP:rtTA) and (2) the tetracycline operator controlling the murine uPA cDNA (tet[O]:muPA). Mice were treated with doxycycline beginning at age 6 wk to initiate uPA overexpression. Single transgenic and wild-type animals served as controls. A second group of double transgenic and control animals were maintained off of doxycycline. At ages 10, 18, and 30 wk, the mice underwent measurements of alveolar size, lung compliance, and total lung capacity. We found that, although the uPA overexpressing mice demonstrated an emphysema phenotype, similar abnormalities occurred in the CCSP-rtTA control animals. These CCSP-rtTA-related alterations occurred even without doxycycline exposure. Evaluation of a second transgenic line possessing the human surfactant protein C promoter controlling rtTA expression also exhibited lung abnormalities consistent with emphysema. These findings indicate that pulmonary epithelial expression of rtTA alone causes an emphysema phenotype in mice. Therefore, when using this system to study emphysema pathogenesis, the inclusion of proper controls is essential for accurate data interpretation.


Assuntos
Enfisema/genética , Expressão Gênica/efeitos dos fármacos , Tetraciclina/metabolismo , Transativadores/genética , Animais , Peso Corporal/genética , Doxiciclina/farmacologia , Genótipo , Cinética , Complacência Pulmonar/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Alvéolos Pulmonares/citologia , Proteína C Associada a Surfactante Pulmonar/genética , Capacidade Pulmonar Total/efeitos dos fármacos , Ativador de Plasminogênio Tipo Uroquinase/genética , Uteroglobina/genética
19.
Am J Respir Cell Mol Biol ; 31(6): 672-8, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15308506

RESUMO

The pathogenesis of pulmonary fibrosis is thought to involve alveolar epithelial injury that, when successfully repaired, can limit subsequent scarring. The plasminogen system participates in this process with the balance between urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1) being a critical determinant of the extent of collagen accumulation that follows lung injury. Because the plasminogen system is known to influence the rate of migration of epithelial cells, including keratinocytes and bronchial epithelial cells, we hypothesized that the balance of uPA and PAI-1 would affect the efficiency of alveolar epithelial cell (AEC) wound repair. Using an in vitro model of AEC wounding, we show that the efficiency of repair is adversely affected by a deficiency in uPA or by the exogenous administration of PAI-1. By using PAI-1 variants and AEC from mice transgenically deficient in vitronectin (Vn), we demonstrate that the PAI-1 effect requires its Vn-binding activity. Furthermore, we have found that cell motility is enhanced by the availability of Vn in the matrix and that the AEC-Vn interaction is mediated, in part, by the alpha(v)beta(1) integrin. The significant effect of uPA and PAI-1 on epithelial repair suggests a mechanism by which the plasminogen system may modulate pulmonary fibrosis.


Assuntos
Bleomicina/análogos & derivados , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Vitronectina/metabolismo , Cicatrização/efeitos dos fármacos , Animais , Bleomicina/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Colágeno/metabolismo , Humanos , Camundongos , Camundongos Knockout , Ligação Proteica , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Vitronectina/deficiência , Vitronectina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...