Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Clin Med ; 12(17)2023 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-37685503

RESUMO

Uterine leiomyomas or uterine fibroids are the most common benign soft tissue tumor in reproductive-aged women. Fumarate hydratase deficient (FH-d) uterine fibroids are a rare subtype that is diagnosed only on pathologic evaluation. FH-d uterine fibroids may be the first indicator of hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome. Therefore, identifying and understanding the clinical implication and diagnosis of FH-d uterine fibroids is critical for early diagnosis of HLRCC. This case series investigates the uncommon yet significant condition of FH-d uterine fibroids. We examined the clinical manifestation, diagnostic imaging, and histopathological characteristics of FH-d uterine fibroids in five cases identified at our institution over the last ten years. All diagnoses were confirmed by pathologic evaluation after surgical treatment. Gynecologists and pathologists play a critical role in the early diagnosis of FH-d uterine fibroids and must recognize the relevant clinical and pathologic findings that raise suspicion about this diagnosis. The detection of these cases is largely dependent on the pathologist's ability to recognize unique histopathologic features. Once these characteristics are identified, it should prompt a referral to a gynecologist to consider conducting germline genetic testing. The management of FH-d uterine fibroids necessitates a multidisciplinary approach, including proper genetic screening and regular surveillance, especially for renal tumors.

3.
Med Sci Educ ; 31(2): 599-606, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34457914

RESUMO

PURPOSE: To assess obstetrician-gynecologist (Ob/Gyn) resident experiences with and preferences for lesbian, gay, bisexual, transgender, and queer (LGBTQ) healthcare training. METHODS: A cross-sectional, web-based survey was deployed to residents from accredited Illinois Ob/Gyn training programs. The survey included 32 questions on resident demographics, LGBTQ training, and self-perceived preparedness in providing LGBTQ patient care. RESULTS: Of 257 eligible Ob/Gyn residents, 105 (41%) responded. Fifty percent of residents felt unprepared to care for lesbian or bisexual patients and 76% felt unprepared to care for transgender patients. Feeling prepared to provide care for lesbian or bisexual patients was associated with attending a university-based program, working in a hospital without religious affiliation, and year of training. Feeling prepared to provide healthcare for transgender patients correlated with grand rounds focused on LGBTQ health and supervised clinical involvement. Regarding training, 62% and 63% of participants stated their programs dedicate 1-5 h per year to lesbian/bisexual healthcare and transgender healthcare training, respectively. Concurrently, 92% desired more education on how to provide healthcare to LGBTQ patients. Perceived barriers to receiving training in LGBTQ healthcare included curricular crowding (85%) and lack of experienced faculty (91%). CONCLUSION: Our assessment indicates Illinois Ob/Gyn residents feel inadequately prepared to address healthcare needs of LGBTQ patients. Although barriers exist, residents desire more education and training in providing healthcare to the LGBTQ community. Future work is needed to address this gap through curricular development to ensure that Ob/Gyn residency graduates are prepared care for LGBTQ patients.

4.
Cancer Epidemiol Biomarkers Prev ; 30(10): 1826-1833, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34272263

RESUMO

BACKGROUND: The influence of prenatal diethylstilbestrol (DES) exposure on cancer incidence among middle-aged men has not been well-characterized. We investigated whether exposure to DES before birth impacts overall cancer risk, and risk of site-specific cancers. METHODS: Men (mean age in 2016 = 62.0 years) who were or were not prenatally DES exposed were identified between 1953 and 1994 and followed for cancer primarily via questionnaire approximately every 5 years between 1994 and 2016. The overall and site-specific cancer rates of the two groups were compared using Poisson regression and proportional hazards modeling with adjustment for age. RESULTS: DES exposure was not associated with either overall cancer [hazard ratio (HR), 0.94; 95% confidence interval (CI), 0.77-1.15] or total prostate cancer rates (HR, 0.95; 95% CI, 0.68-1.33), but was inversely associated with urinary tract cancer incidence (HR, 0.48; 95% CI, 0.23-1.00). CONCLUSIONS: There was no increase in either overall or prostate cancer rates among men prenatally DES exposed relative to those unexposed. An unexpected risk reduction was observed for urinary system cancers among the exposed relative to those unexposed. These findings suggest that prenatal DES exposure is unlikely to be an important contributor to cancer development in middle-aged men. IMPACT: The results of this study could lend reassurance to middle-aged men who were prenatally DES exposed that their exposure does not adversely influence their overall cancer risk.


Assuntos
Neoplasias , Efeitos Tardios da Exposição Pré-Natal , Dietilestilbestrol/efeitos adversos , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Neoplasias/induzido quimicamente , Neoplasias/epidemiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Risco
5.
Cancer Res ; 80(8): 1748-1761, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32054768

RESUMO

Adipocytes are critical for ovarian cancer cells to home to the omentum, but the metabolic changes initiated by this interaction are unknown. To this end, we carried out unbiased mass spectrometry-based metabolomic and proteomic profiling of cancer cells cocultured with primary human omental adipocytes. Cancer cells underwent significant proteo-metabolomic alteration(s), typified by changes in the lipidome with corresponding upregulation of lipid metabolism proteins. FABP4, a lipid chaperone protein, was identified as the critical regulator of lipid responses in ovarian cancer cells cocultured with adipocytes. Subsequently, knockdown of FABP4 resulted in increased 5-hydroxymethylcytosine levels in the DNA, downregulation of gene signatures associated with ovarian cancer metastasis, and reduced clonogenic cancer cell survival. In addition, clustered regularly interspaced short palindromic repeats (CRISPR)-mediated knockout of FABP4 in high-grade serous ovarian cancer cells reduced metastatic tumor burden in mice. Consequently, a small-molecule inhibitor of FABP4 (BMS309403) not only significantly reduced tumor burden in a syngeneic orthotopic mouse model but also increased the sensitivity of cancer cells toward carboplatin both in vitro and in vivo. Taken together, these results show that targeting FABP4 in ovarian cancer cells can inhibit their ability to adapt and colonize lipid-rich tumor microenvironments, providing an opportunity for specific metabolic targeting of ovarian cancer metastasis. SIGNIFICANCE: Ovarian cancer metastatic progression can be restricted by targeting a critical regulator of lipid responses, FABP4.


Assuntos
Adipócitos/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proteínas de Ligação a Ácido Graxo/metabolismo , Neoplasias Ovarianas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/análise , Animais , Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Carboplatina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Técnicas de Cocultura , Metilação de DNA , Regulação para Baixo , Proteínas de Ligação a Ácido Graxo/antagonistas & inibidores , Proteínas de Ligação a Ácido Graxo/genética , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Metabolismo dos Lipídeos , Lipidômica , Espectrometria de Massas , Metabolômica/métodos , Camundongos , Camundongos Nus , Metástase Neoplásica/prevenção & controle , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Omento/citologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Análise Serial de Proteínas , Proteômica/métodos , Pirazóis/farmacologia , Carga Tumoral/efeitos dos fármacos , Regulação para Cima
6.
Cell Rep ; 29(12): 4086-4098.e6, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31851935

RESUMO

The tumor microenvironment (TME) plays a pivotal role in cancer progression, and, in ovarian cancer (OvCa), the primary TME is the omentum. Here, we show that the diabetes drug metformin alters mesothelial cells in the omental microenvironment. Metformin interrupts bidirectional signaling between tumor and mesothelial cells by blocking OvCa cell TGF-ß signaling and mesothelial cell production of CCL2 and IL-8. Inhibition of tumor-stromal crosstalk by metformin is caused by the reduced expression of the tricarboxylic acid (TCA) enzyme succinyl CoA ligase (SUCLG2). Through repressing this TCA enzyme and its metabolite, succinate, metformin activated prolyl hydroxylases (PHDs), resulting in the degradation of hypoxia-inducible factor 1α (HIF1α) in mesothelial cells. Disruption of HIF1α-driven IL-8 signaling in mesothelial cells by metformin results in reduced OvCa invasion in an organotypic 3D model. These findings indicate that tumor-promoting signaling between mesothelial and OvCa cells in the TME can be targeted using metformin.


Assuntos
Carcinogênese/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Metformina/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Células Estromais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Feminino , Humanos , Hipoglicemiantes/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Prolil Hidroxilases/genética , Prolil Hidroxilases/metabolismo , Células Estromais/patologia , Succinato-CoA Ligases/genética , Succinato-CoA Ligases/metabolismo , Células Tumorais Cultivadas
7.
Mol Cancer Res ; 17(4): 870-881, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30655321

RESUMO

The role of phospholipid signaling in ovarian cancer is poorly understood. Sphingosine-1-phosphate (S1P) is a bioactive metabolite of sphingosine that has been associated with tumor progression through enhanced cell proliferation and motility. Similarly, sphingosine kinases (SPHK), which catalyze the formation of S1P and thus regulate the sphingolipid rheostat, have been reported to promote tumor growth in a variety of cancers. The findings reported here show that exogenous S1P or overexpression of SPHK1 increased proliferation, migration, invasion, and stem-like phenotypes in ovarian cancer cell lines. Likewise, overexpression of SPHK1 markedly enhanced tumor growth in a xenograft model of ovarian cancer, which was associated with elevation of key markers of proliferation and stemness. The diabetes drug, metformin, has been shown to have anticancer effects. Here, we found that ovarian cancer patients taking metformin had significantly reduced serum S1P levels, a finding that was recapitulated when ovarian cancer cells were treated with metformin and analyzed by lipidomics. These findings suggested that in cancer the sphingolipid rheostat may be a novel metabolic target of metformin. In support of this, metformin blocked hypoxia-induced SPHK1, which was associated with inhibited nuclear translocation and transcriptional activity of hypoxia-inducible factors (HIF1α and HIF2α). Further, ovarian cancer cells with high SPHK1 were found to be highly sensitive to the cytotoxic effects of metformin, whereas ovarian cancer cells with low SPHK1 were resistant. Together, the findings reported here show that hypoxia-induced SPHK1 expression and downstream S1P signaling promote ovarian cancer progression and that tumors with high expression of SPHK1 or S1P levels might have increased sensitivity to the cytotoxic effects of metformin. IMPLICATIONS: Metformin targets sphingolipid metabolism through inhibiting SPHK1, thereby impeding ovarian cancer cell migration, proliferation, and self-renewal.


Assuntos
Metformina/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Movimento Celular/efeitos dos fármacos , Feminino , Humanos , Hipoglicemiantes/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Clin Cancer Res ; 25(6): 1786-1794, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30154229

RESUMO

PURPOSE: To establish a cohort of high-risk women undergoing intensive surveillance for breast cancer.Experimental Design: We performed dynamic contrast-enhanced MRI every 6 months in conjunction with annual mammography (MG). Eligible participants had a cumulative lifetime breast cancer risk ≥20% and/or tested positive for a pathogenic mutation in a known breast cancer susceptibility gene. RESULTS: Between 2004 and 2016, we prospectively enrolled 295 women, including 157 mutation carriers (75 BRCA1, 61 BRCA2); participants' mean age at entry was 43.3 years. Seventeen cancers were later diagnosed: 4 ductal carcinoma in situ (DCIS) and 13 early-stage invasive breast cancers. Fifteen cancers occurred in mutation carriers (11 BRCA1, 3 BRCA2, 1 CDH1). Median size of the invasive cancers was 0.61 cm. No patients had lymph node metastasis at time of diagnosis, and no interval invasive cancers occurred. The sensitivity of biannual MRI alone was 88.2% and annual MG plus biannual MRI was 94.1%. The cancer detection rate of biannual MRI alone was 0.7% per 100 screening episodes, which is similar to the cancer detection rate of 0.7% per 100 screening episodes for annual MG plus biannual MRI. The number of recalls and biopsies needed to detect one cancer by biannual MRI were 2.8 and 1.7 in BRCA1 carriers, 12.0 and 8.0 in BRCA2 carriers, and 11.7 and 5.0 in non-BRCA1/2 carriers, respectively. CONCLUSIONS: Biannual MRI performed well for early detection of invasive breast cancer in genomically stratified high-risk women. No benefit was associated with annual MG screening plus biannual MRI screening.See related commentary by Kuhl and Schrading, p. 1693.


Assuntos
Proteína BRCA1/genética , Neoplasias da Mama/diagnóstico , Detecção Precoce de Câncer/métodos , Imageamento por Ressonância Magnética/métodos , Programas de Rastreamento/métodos , Adulto , Biópsia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Predisposição Genética para Doença , Humanos , Mamografia , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Estudos Prospectivos
9.
Artigo em Inglês | MEDLINE | ID: mdl-31517176

RESUMO

PURPOSE: To describe a snapshot of international genetic testing practices, specifically regarding the use of multigene panels, for hereditary breast/ovarian cancers. We conducted a survey through the Evidence-Based Network for the Interpretation of Germline Mutant Alleles (ENIGMA) consortium, covering questions about 16 non-BRCA1/2 genes. METHODS: Data were collected via in-person and paper/electronic surveys. ENIGMA members from around the world were invited to participate. Additional information was collected via country networks in the United Kingdom and in Italy. RESULTS: Responses from 61 cancer genetics practices across 20 countries showed that 16 genes were tested by > 50% of the centers, but only six (PALB2, TP53, PTEN, CHEK2, ATM, and BRIP1) were tested regularly. US centers tested the genes most often, whereas United Kingdom and Italian centers with no direct ENIGMA affiliation at the time of the survey were the least likely to regularly test them. Most centers tested the 16 genes through multigene panels; some centers tested TP53, PTEN, and other cancer syndrome-associated genes individually. Most centers reported (likely) pathogenic variants to patients and would test family members for such variants. Gene-specific guidelines for breast and ovarian cancer risk management were limited and differed among countries, especially with regard to starting age and type of imaging and risk-reducing surgery recommendations. CONCLUSION: Currently, a small number of genes beyond BRCA1/2 are routinely analyzed worldwide, and management guidelines are limited and largely based on expert opinion. To attain clinical implementation of multigene panel testing through evidence-based management practices, it is paramount that clinicians (and patients) participate in international initiatives that share panel testing data, interpret sequence variants, and collect prospective data to underpin risk estimates and evaluate the outcome of risk intervention strategies.

11.
Semin Reprod Med ; 35(5): 420-425, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-29073680

RESUMO

The constructs and the provision of preconception and obstetrical care have historically been based on the assumption of heterosexuality, and have often excluded lesbian women. However, due to significant strides in lesbian, gay, bisexual, transgender, and queer (LGBTQ) civil rights, more lesbian women desire to create and expand their families, and lesbian parented families are increasing. This places obstetrical care providers at the forefront of the movement to build inclusive health care environments. Therefore, it is incumbent upon those of us who work in obstetrics to understand, recognize, and respect the unique cultural considerations that pertain to lesbian women and couples seeking parenthood. This review seeks to provide culturally sensitive guidance on the specific concerns and challenges lesbians face, from preconception care to postpartum care, and briefly addresses legal issues and considerations for the nonbiologic mother. The recommendations outlined here are drawn from studies of the experiences of lesbian women with pregnancy. However, the scientific literature is very limited, and there is a clear need for additional obstetrical research focused on this patient group. As professionals committed to assuring optimal outcomes for all obstetrical patients, it is crucial that we promote the inclusion of sexual minority women in our clinical practices and research endeavors.


Assuntos
Assistência à Saúde Culturalmente Competente , Fertilização in vitro , Homossexualidade Feminina , Casamento , Serviços de Saúde Materna , Pais , Minorias Sexuais e de Gênero , Cônjuges , Assistência à Saúde Culturalmente Competente/legislação & jurisprudência , Assistência à Saúde Culturalmente Competente/normas , Feminino , Fertilização in vitro/legislação & jurisprudência , Fertilização in vitro/normas , Conhecimentos, Atitudes e Prática em Saúde , Política de Saúde , Homossexualidade Feminina/psicologia , Humanos , Casamento/legislação & jurisprudência , Casamento/psicologia , Serviços de Saúde Materna/legislação & jurisprudência , Serviços de Saúde Materna/normas , Pais/psicologia , Formulação de Políticas , Gravidez , Minorias Sexuais e de Gênero/legislação & jurisprudência , Minorias Sexuais e de Gênero/psicologia , Cônjuges/legislação & jurisprudência , Cônjuges/psicologia , Resultado do Tratamento
12.
Cancer Prev Res (Phila) ; 10(4): 255-266, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28264838

RESUMO

Mutations in the breast cancer susceptibility gene 1 (BRCA1) are associated with an increased risk of developing epithelial ovarian cancer. However, beyond the role of BRCA1 in DNA repair, little is known about other mechanisms by which BRCA1 impairment promotes carcinogenesis. Given that altered metabolism is now recognized as important in the initiation and progression of cancer, we asked whether the loss of BRCA1 changes metabolism in the cells of origin of ovarian cancer. The findings show that silencing BRCA1 in ovarian surface epithelial and fallopian tube cells increased glycolysis. Furthermore, when these cells were transfected with plasmids carrying deleterious BRCA1 mutations (5382insC or the P1749R), there was an increase in hexokinase-2 (HK2), a key glycolytic enzyme. This effect was mediated by MYC and the STAT3. To target the metabolic phenotype induced by loss of BRCA1, a drug-repurposing approach was used and aspirin was identified as an agent that counteracted the increase in HK2 and the increase in glycolysis induced by BRCA1 impairment. Evidence from this study indicates that the tumor suppressor functions of BRCA1 extend beyond DNA repair to include metabolic endpoints and identifies aspirin as an ovarian cancer chemopreventive agent capable of reversing the metabolic derangements caused by loss of BRCA1. Cancer Prev Res; 10(4); 255-66. ©2017 AACR.


Assuntos
Proteína BRCA1/genética , Células Epiteliais/metabolismo , Glicólise , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Aspirina/farmacologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Quimioprevenção/métodos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Tubas Uterinas/metabolismo , Feminino , Glicólise/efeitos dos fármacos , Humanos , Mutação
13.
Cell Metab ; 24(5): 728-739, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27746051

RESUMO

Repurposing metformin for cancer therapy is attractive due to its safety profile, epidemiological evidence, and encouraging data from human clinical trials. Although it is known to systemically affect glucose metabolism in liver, muscle, gut, and other tissues, the molecular determinants that predict a patient response in cancer remain unknown. Here, we carry out an integrative metabolomics analysis of metformin action in ovarian cancer. Metformin accumulated in patient biopsies, and pathways involving nucleotide metabolism, redox, and energy status, all related to mitochondrial metabolism, were affected in treated tumors. Strikingly, a metabolic signature obtained from a patient with an exceptional clinical outcome mirrored that of a responsive animal tumor. Mechanistically, we demonstrate with stable isotope tracing that these metabolic signatures are due to an inability to adapt nutrient utilization in the mitochondria. This analysis provides new insights into mitochondrial metabolism and may lead to more precise indications of metformin in cancer.


Assuntos
Carbono/metabolismo , Metformina/farmacologia , Mitocôndrias/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Glucose/farmacologia , Humanos , Metaboloma/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Neoplasias Ovarianas/patologia , Especificidade por Substrato/efeitos dos fármacos , Resultado do Tratamento
14.
PLoS One ; 11(1): e0147145, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26788855

RESUMO

There is increasing evidence that metformin, a commonly used treatment for diabetes, might have the potential to be repurposed as an economical and safe cancer therapeutic. The aim of this study was to determine whether stage III-IV or recurrent endometrial cancer patients who are using metformin during treatment with chemotherapy have improved survival. To test this we analyzed a retrospective cohort of subjects at two independent institutions who received chemotherapy for stage III-IV or recurrent endometrial cancer from 1992 to 2011. Diagnosis of diabetes, metformin use, demographics, endometrial cancer clinico-pathologic parameters, and survival duration were abstracted. The primary outcome was overall survival. The final cohort included 349 patients, 31 (8.9%) had diabetes and used metformin, 28 (8.0%) had diabetes but did not use metformin, and 291 (83.4%) did not have diabetes. The results demonstrate that the median overall survival was 45.6 months for patients with diabetes who used metformin compared to 12.5 months for patients with diabetes who did not use metformin and 28.5 months for patients without diabetes (log-rank test comparing the three groups P = 0.006). In a model adjusted for confounders, the difference in survival between the three groups remained statistically significant (P = 0.023). The improvement in survival among metformin users was not explained by better baseline health status or more aggressive use of chemotherapy. Overall, the findings in this retrospective cohort of endometrial cancer patients with stage III-IV or recurrent disease treated with chemotherapy indicate that patients with diabetes who were concurrently treated with metformin survived longer than patients with diabetes who did not use metformin.


Assuntos
Adenocarcinoma de Células Claras/tratamento farmacológico , Carcinossarcoma/tratamento farmacológico , Cistadenocarcinoma Seroso/tratamento farmacológico , Neoplasias do Endométrio/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Adenocarcinoma de Células Claras/mortalidade , Adenocarcinoma de Células Claras/patologia , Idoso , Carcinossarcoma/mortalidade , Carcinossarcoma/patologia , Cistadenocarcinoma Seroso/mortalidade , Cistadenocarcinoma Seroso/patologia , Neoplasias do Endométrio/mortalidade , Neoplasias do Endométrio/patologia , Feminino , Seguimentos , Humanos , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida
15.
Nat Rev Cancer ; 15(11): 668-79, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26493647

RESUMO

High-grade serous ovarian cancer (HGSOC) accounts for 70-80% of ovarian cancer deaths, and overall survival has not changed significantly for several decades. In this Opinion article, we outline a set of research priorities that we believe will reduce incidence and improve outcomes for women with this disease. This 'roadmap' for HGSOC was determined after extensive discussions at an Ovarian Cancer Action meeting in January 2015.


Assuntos
Cistadenocarcinoma Seroso/mortalidade , Cistadenocarcinoma Seroso/prevenção & controle , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/prevenção & controle , Cistadenocarcinoma Seroso/patologia , Feminino , Humanos , Gradação de Tumores , Neoplasias Ovarianas/patologia , Prognóstico , Taxa de Sobrevida
16.
Oncotarget ; 6(27): 23548-60, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26172303

RESUMO

Increasing interest in repurposing the diabetic medication metformin for cancer treatment has raised important questions about the translation of promising preclinical findings to therapeutic efficacy, especially in non-diabetic patients. A significant limitation of the findings to date is the use of supraphysiologic metformin doses and hyperglycemic conditions in vitro. Our goals were to determine the impact of hyperglycemia on metformin response and to address the applicability of metformin as a cancer therapeutic in non-diabetic patients. In normoglycemic conditions, lower concentrations of metformin were required to inhibit cell viability, while metformin treatment in hyperglycemic conditions resulted in increased glucose uptake and glycolytic flux, contributing to cell survival. Mechanistically, maintenance of c-Myc expression under conditions of hyperglycemia or via gene amplification facilitated metabolic escape from the effects of metformin. In vivo, treatment of an ovarian cancer mouse model with metformin resulted in greater tumor weight reduction in normoglycemic vs. hyperglycemic mice, with increased c-Myc expression observed in metformin-treated hyperglycemic mice. These findings indicate that hyperglycemia inhibits the anti-cancer effects of metformin in vitro and in vivo. Furthermore, our results suggest that metformin may elicit stronger responses in normoglycemic vs. hyperglycemic patients, highlighting the need for prospective clinical testing in patients without diabetes.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hiperglicemia/metabolismo , Metformina/química , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Animais , Antineoplásicos/química , Ascite/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Glucose/metabolismo , Glicólise , Humanos , Hipoglicemiantes/química , Ácido Láctico/química , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/metabolismo , Via de Pentose Fosfato , Fenformin/química , Proteínas Proto-Oncogênicas c-myc/metabolismo
17.
Clin Cancer Res ; 21(4): 680-6, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25691772

RESUMO

A model of tumor metabolism is proposed that describes how the complementary metabolic functions of the local stroma and the tumor cells contribute to cancer progression. Cancer cells alter the metabolism of cancer-associated fibroblasts to obtain lactate and amino acids, which are utilized for energy production, rapid growth, and resistance to chemotherapy drugs. Cancer cells use glutamine supplied by cancer-associated fibroblasts to replenish tricarboxylic acid cycle intermediates and as a nitrogen source for nucleotide synthesis. Moreover, adipocytes in the microenvironment attract cancer cells through the secretion of inflammatory cytokines and proteases. The cancer cells then induce metabolic changes in the adipocytes to acquire free fatty acids that are oxidized by cancer cells to generate energy for proliferation. Increasing knowledge about the metabolic symbiosis within the tumor has led to novel therapeutic strategies designed to restrict metabolic adaptation, including inhibiting lactate transporters and repurposing antidiabetic drugs (thiazolidinediones, metformin).


Assuntos
Modelos Biológicos , Neoplasias/metabolismo , Transdução de Sinais/fisiologia , Microambiente Tumoral/fisiologia , Humanos , Estresse Oxidativo/fisiologia
18.
Am J Obstet Gynecol ; 212(4): 479.e1-479.e10, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25446664

RESUMO

OBJECTIVE: There is increasing preclinical evidence indicating that metformin, a medication commonly used for type 2 diabetes mellitus, may protect against cancer. Motivated by this emerging evidence we asked 2 questions: (1) can metformin prevent ovarian cancer growth by altering metabolism and (2) will metformin increase sensitivity to chemotherapy. STUDY DESIGN: The effect of metformin in ovarian cancer was tested in vitro and with 2 different mouse models. In vitro, cell lines (n = 6) were treated with metformin (10-40 mmol/L) or phosphate-buffered saline solution and cellular proliferation and metabolic alterations (adenosine monophosphate-activated protein kinase activity, glycolysis, and lipid synthesis) were compared between the 2 groups. In mouse models, a prevention study was performed by treating mice with metformin (250 mg/kg/d intraperitoneally) or placebo for 2 weeks followed by intraperitoneal injection of the SKOV3ip1 human ovarian cancer cell line, and the mean number of tumor implants in each treatment group was compared. In a treatment study, the LSL-K-ras(G12D/+)/PTEN(floxP/floxP) genetic mouse model of ovarian cancer was used. Mice were treated with placebo, paclitaxel (3 mg/kg/wk intraperitoneally for 7 weeks), metformin (100 mg/kg/d in water for 7 weeks), or paclitaxel plus metformin, and tumor volume was compared among treatment groups. RESULTS: In vitro, metformin decreased proliferation of ovarian cancer cell lines and induced cell cycle arrest, but not apoptosis. Further analysis showed that metformin altered several aspects of metabolism including adenosine monophosphate-activated protein kinase activity, glycolysis, and lipid synthesis. In the prevention mouse model, mice that were pretreated with metformin had 60% fewer tumor implants compared with controls (P < .005). In the treatment study, mice that were treated with paclitaxel plus metformin had a 60% reduction in tumor weight compared with controls (P = .02), which is a level of tumor reduction greater than that resulting from either paclitaxel or metformin alone. CONCLUSION: Based on these results, we conclude that metformin alters metabolism in ovarian cancer cells, prevents tumor growth, and increases sensitivity to chemotherapy in vitro and in mouse models. These preclinical findings suggest that metformin warrants further investigation for use as an ovarian cancer therapeutic.


Assuntos
Antineoplásicos/uso terapêutico , Metformina/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/prevenção & controle , Paclitaxel/uso terapêutico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Metformina/farmacologia , Camundongos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carga Tumoral/efeitos dos fármacos
19.
Gynecol Oncol ; 135(3): 614-21, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25455733

RESUMO

OBJECTIVE: There is increasing pre-clinical and clinical evidence that metformin, a commonly used diabetes medication, has a protective effect in cancer. The aim of this review is to discuss metformin's anti-cancer molecular mechanisms of action and to summarize the current literature demonstrating metformin's potential in gynecologic cancer prevention and treatment. METHODS: A PubMed search was conducted combining the keywords "metformin" with "neoplasm", "uterine neoplasms", "ovarian neoplasms", and "uterine cervical neoplasms". Studies published in English between 1994 and 2014 were included. RESULTS: Pre-clinical studies in endometrial, ovarian, and cervical cancer suggest that metformin inhibits the growth of cancer cells. The primary molecular mechanism mediating this effect appears to be the activation of AMP-activated protein kinase (AMPK) and the subsequent inhibition of mammalian targets of rapamycin (mTOR). The pre-clinical findings are augmented by clinical studies indicating that metformin use is associated with a reduced risk of cancer and improved survival in diabetic women with ovarian and endometrial cancers. No clinical analyses have evaluated metformin use and cervical cancer. Overall, the data showing a favorable effect of metformin is strongest for endometrial and ovarian cancer and prospective clinical testing is ongoing in these two malignancies. CONCLUSIONS: Numerous clinical studies have reported an association between metformin use by diabetic patients and improved outcomes in gynecologic cancers. In addition, pre-clinical reports have identified plausible biological mechanisms to explain the molecular mechanism of action of metformin in cancer. However, the most important question remains unanswered: Will metformin be effective against cancer in patients without diabetes? Until this question is answered with prospective clinical testing, the role of metformin in the treatment or prevention of gynecologic malignancies remains theoretical and the clinical use of metformin as a cancer therapeutic is experimental.


Assuntos
Neoplasias dos Genitais Femininos/prevenção & controle , Metformina/administração & dosagem , Feminino , Humanos
20.
J Clin Invest ; 124(10): 4614-28, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25202979

RESUMO

Ovarian cancer (OvCa) metastasizes to organs in the abdominal cavity, such as the omentum, which are covered by a single layer of mesothelial cells. Mesothelial cells are generally thought to be "bystanders" to the metastatic process and simply displaced by OvCa cells to access the submesothelial extracellular matrix. Here, using organotypic 3D cultures, we found that primary human mesothelial cells secrete fibronectin in the presence of OvCa cells. Moreover, we evaluated the tumor stroma of 108 human omental metastases and determined that fibronectin was consistently overexpressed in these patients. Blocking fibronectin production in primary mesothelial cells in vitro or in murine models, either genetically (fibronectin 1 floxed mouse model) or via siRNA, decreased adhesion, invasion, proliferation, and metastasis of OvCa cells. Using a coculture model, we determined that OvCa cells secrete TGF-ß1, which in turn activates a TGF-ß receptor/RAC1/SMAD-dependent signaling pathway in the mesothelial cells that promotes a mesenchymal phenotype and transcriptional upregulation of fibronectin. Additionally, blocking α5 or ß1 integrin function with antibodies reduced metastasis in an orthotopic preclinical model of OvCa metastasis. These findings indicate that cancer-associated mesothelial cells promote colonization during the initial steps of OvCa metastasis and suggest that mesothelial cells actively contribute to metastasis.


Assuntos
Células Epiteliais/citologia , Fibronectinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/patologia , Animais , Adesão Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Matriz Extracelular/metabolismo , Feminino , Humanos , Integrinas/metabolismo , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias Ovarianas/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...