Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Pathol ; 262(2): 129-136, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38013631

RESUMO

Trastuzumab has demonstrated clinical efficacy in the treatment of HER2-positive serous endometrial cancer (EC), which led to its incorporation into standard-of-care management of this aggressive disease. Acquired resistance remains an important challenge, however, and its underlying mechanisms in EC are unknown. To define the molecular changes that occur in response to anti-HER2 therapy in EC, targeted next-generation sequencing (NGS), HER2 immunohistochemistry (IHC), and fluorescence in situ hybridization (FISH) were performed on pre- and post-treatment tumour samples from 14 patients with EC treated with trastuzumab or trastuzumab emtansine. Recurrent tumours after anti-HER2 therapy acquired additional genetic alterations compared with matched pre-treatment ECs and frequently showed decreased HER2 protein expression by IHC (7/14, 50%). Complete/near-complete absence of HER2 protein expression (score 0/1+) observed post-treatment (4/14, 29%) was associated with retained HER2 gene amplification (n = 3) or copy number neutral status (n = 1). Whole-exome sequencing performed on primary and recurrent tumours from the latter case, which exhibited genetic heterogeneity of HER2 amplification in the primary tumour, revealed selection of an early HER2-non-amplified clone following therapy. Our findings demonstrate that loss of target expression, by selection of HER2-non-amplified clones or, more commonly, by downregulation of expression, may constitute a mechanism of resistance to anti-HER2 therapy in HER2-positive EC. © 2023 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neoplasias do Endométrio , Receptor ErbB-2 , Feminino , Humanos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Hibridização in Situ Fluorescente , Recidiva Local de Neoplasia/genética , Trastuzumab/uso terapêutico , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Amplificação de Genes
2.
Trans Am Clin Climatol Assoc ; 133: 274-282, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37701602

RESUMO

Endometrial carcinoma is the most common malignancy of the genital tract in females in the United States, and it is one of the few human cancers increasing in incidence and mortality. Numerous studies have found an association of endometrial carcinoma with obesity, diabetes, and unopposed estrogen stimulation. Molecular studies, in our lab and others, have shown that endometrial carcinoma has a high frequency of alterations in the Phosphoinositide 3-kinase (PIK3) pathway, and notably, coexisting abnormalities in more than one member of the pathway are common. We have combined studies on primary human tumors and genetic mouse models to explore the role of the PIK3 pathway and estrogen, and their interactions, in the development and progression of endometrial carcinoma. Abnormalities in the PIK3 pathway do not simply play redundant roles in endometrial carcinoma and, although not required, the presence of estrogen can alter the course of the disease.


Assuntos
Neoplasias do Endométrio , Fosfatidilinositol 3-Quinases , Feminino , Humanos , Animais , Camundongos , Neoplasias do Endométrio/genética , Estrogênios
3.
Int J Gynecol Pathol ; 41(3): 207-219, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34483300

RESUMO

Low-grade, low-stage endometrioid carcinomas (LGLS EC) demonstrate 5-yr survival rates up to 95%. However, a small subset of these tumors recur, and little is known about prognostic markers or established mutation profiles associated with recurrence. The goal of the current study was to identify the molecular profiles of the primary carcinomas and the genomic differences between primary tumors and subsequent recurrences. Four cases of LGLS EC with recurrence and 8 cases without recurrence were evaluated via whole-exome sequencing. Three of the 4 recurrent tumors were evaluated via Oncomine Comprehensive Assay. The resulting molecular profiles of the primary and recurrent tumors were compared. Two of the 3 recurrent cases showed additional mutations in the recurrence. One recurrent tumor included an additional TP53 mutation and the other recurrent tumor showed POLE and DDR2 kinase gene mutation. The POLE mutation occurred outside the exonuclease domain. PIK3CA mutations were detected in 4 of 4 primary LGLS EC with recurrence and in 3 of 8 disease-free cases. LGLS EC with recurrence showed higher MSIsensor scores compared with LGLS without recurrence. The level of copy number gains in LGLS EC with recurrence was larger than LGLS EC without recurrence. This pilot study showed 1 of 3 recurrent cases gained a mutation associated with genetic instability (TP53) and 1 of them also acquired a mutation in the DDR2 kinase, a potential therapeutic target. We also noted a higher level of copy number gains, MSIsensor scores and PIK3CA mutations in the primary tumors that later recurred.


Assuntos
Carcinoma Endometrioide , Receptor com Domínio Discoidina 2 , Neoplasias do Endométrio , Carcinoma Endometrioide/diagnóstico , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patologia , Classe I de Fosfatidilinositol 3-Quinases/genética , Receptor com Domínio Discoidina 2/genética , Neoplasias do Endométrio/diagnóstico , Neoplasias do Endométrio/genética , Feminino , Humanos , Mutação , Projetos Piloto
4.
Int J Gynecol Pathol ; 37(5): 405-413, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29019871

RESUMO

Historically, endometrial carcinomas have been classified primarily according to their histology. However, the use of immunohistochemistry has become commonplace in their evaluation, particularly in diagnostically challenging cases. Our objective was to evaluate mixed endometrial carcinomas using a well-established panel of biomarkers to assess the consistency and utility of these stains in clinical diagnosis. Eighteen cases comprised of various combinations of classical serous (SC), endometrioid (EC), and clear cell (CC) morphologies were identified and subjected to a panel of immunohistochemical markers including p53, p16, Ki67, estrogen receptor, progesterone receptor, and Napsin A. Intensity and extent of staining were evaluated on 4-tiered and 5-tiered scales, respectively. The typical immunostaining pattern expected for the individual tumor components was seen in only 3 cases, while in 15 cases an unexpected pattern was observed with at least one immunomarker. By tumor type, the most common unexpected finding in EC/SC carcinoma cases was diffuse positivity for p16 and/or estrogen receptor/progesterone receptor in both components, while in SC/CC, diffuse positivity for p53 in both components was most frequently seen, and in SC/CC/EC, Napsin A negativity was most commonly observed. Despite displaying diagnostic morphology, components of many mixed endometrial carcinomas may not exhibit expected immunohistochemical features. This may be due to the fact that these carcinomas arise from a single clone with subsequent divergence, resulting in a tumor with both mixed histologic and genetic features. It is important to note that these tumors may not demonstrate the immunohistochemical prototype of their constituents and should be approached accordingly from a diagnostic perspective.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias do Endométrio/diagnóstico , Neoplasias do Endométrio/patologia , Adenocarcinoma de Células Claras/diagnóstico , Adenocarcinoma de Células Claras/patologia , Idoso , Idoso de 80 Anos ou mais , Carcinoma Endometrioide/diagnóstico , Carcinoma Endometrioide/patologia , Cistadenocarcinoma Seroso/diagnóstico , Cistadenocarcinoma Seroso/patologia , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade
5.
Adv Exp Med Biol ; 943: 261-273, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27910071

RESUMO

The PI3K/PTEN/AKT pathway is the most frequently mutated pathway in endometrial carcinoma. Mouse models are invaluable tools to understand, at the molecular level, the contributions of components of this pathway towards initiation and progression of endometrial carcinoma. This chapter summarizes results of germline and tissue specific knockout mouse models generated to understand how mutations in components of this pathway lead to development of carcinoma and its interactions with other frequently altered pathways like mismatch repair and estrogen signaling. The mouse models show that loss of both alleles of Pten is necessary and sufficient for complex atypical hyperplasia (CAH) to develop but insufficient for progression to carcinoma. Additional events like mutations in Pik3ca or mismatch repair deficiency are required for progression to carcinoma. The models show that the interaction between Pten and estrogen signaling is complex. In the absence of estrogen, Pten loss is sufficient for development of CAH. Additionally, lack of ERα on a background of Pten loss leads to the development of carcinoma.


Assuntos
Neoplasias do Endométrio/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Classe I de Fosfatidilinositol 3-Quinases , Modelos Animais de Doenças , Neoplasias do Endométrio/genética , Feminino , Humanos , Camundongos , Camundongos Knockout , Mutação , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética
6.
Cancer Invest ; 34(3): 148-54, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26986692

RESUMO

Ovarian cancers diagnosed between 2000 and 2013 were examined and cases with and without endometriosis compared. Among 139 epithelial ovarian, there were 49 (35%) with endometriosis and 90 (65%) without endometriosis. Endometriosis associated ovarian cancers were more likely to be confined to the pelvis (54% vs. 9%, p < 0.0001) and lower grade (51% vs. 29%, p = 0.014). Younger age and earlier stage independently predicted the presence of endometriosis (p = 0.0011 and p < 0.0001, respectively). Ovarian cancer patients with endometriosis had improved PFS and OS [(HR = 0.20; 95% CI, 0.09-0.43), (HR = 0.18; 95% CI, 0.04-0.81)], compared to patients without endometriosis; however, endometriosis had no independent prognostic significance.


Assuntos
Endometriose/diagnóstico , Neoplasias Epiteliais e Glandulares/diagnóstico , Neoplasias Ovarianas/diagnóstico , Idoso , Carcinoma Epitelial do Ovário , Intervalo Livre de Doença , Endometriose/mortalidade , Feminino , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Ovarianas/mortalidade , Prognóstico , Modelos de Riscos Proporcionais
7.
Int J Gynecol Cancer ; 24(7): 1262-7, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25078343

RESUMO

OBJECTIVES: Type II endometrial carcinomas-uterine carcinosarcomas or uterine malignant mesodermal mixed tumors (UMMMTs), clear cell carcinomas (UCCs), and uterine serous carcinomas (USCs)-are aggressive malignancies that present with advanced disease and have high mortality rates. PIK3CA mutations are commonly found in endometrial cancers. The objective of the study was to characterize molecular alterations in the PIK3CA gene in these tumors. METHODS: A total of 84 cases (20 UMMMTs, 18 UCCs, and 46 USCs) were selected from the surgical pathology files of Weill Cornell Medical College and Johns Hopkins Hospital. The diagnoses were confirmed by gynecologic pathologists (L.H.E. and A.Y.). DNA was extracted from paraffin-embedded tissue. Polymerase chain reaction was performed for mutational analysis. All the studies were performed in accordance with approved Institutional Review Board protocols. RESULTS: Mutations in the PIK3CA gene were identified in 3 (15%) of 20 UMMMT, 3 (16.7%) of 18 UCC, and 10 (21.7%) of 46 USC cases. We report novel mutations in PIK3CA in uterine carcinosarcoma. CONCLUSIONS: A significant percentage of UMMMTs, UCCs, and USCs have mutations in PIK3CA. Further investigation is needed to develop targeted therapies for these aggressive uterine cancers.


Assuntos
Adenocarcinoma de Células Claras/genética , Carcinossarcoma/genética , Cistadenocarcinoma Seroso/genética , Mutação , Fosfatidilinositol 3-Quinases/genética , Neoplasias Uterinas/genética , Adenocarcinoma de Células Claras/epidemiologia , Adenocarcinoma de Células Claras/patologia , Substituição de Aminoácidos , Carcinossarcoma/epidemiologia , Carcinossarcoma/patologia , Classe I de Fosfatidilinositol 3-Quinases , Cistadenocarcinoma Seroso/epidemiologia , Cistadenocarcinoma Seroso/patologia , Análise Mutacional de DNA , Feminino , Humanos , Polimorfismo de Nucleotídeo Único , Deleção de Sequência , Neoplasias Uterinas/epidemiologia , Neoplasias Uterinas/patologia
8.
Mol Hum Reprod ; 19(7): 463-72, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23475984

RESUMO

Heightened mammalian target of rapamycin complex 1 (mTORC1) activity by genetic deletion of its direct inhibitor, Tsc1, is associated with aberrant development and dysfunction of the female reproductive tract in mice. Here, we compared the phenotypes of mice with conditional deletion of Tsc1 in the female reproductive tract by either progesterone receptor (PR)-Cre (Tsc1(PR(d/d))), which inactivates Tsc1 in all major cell types in the uterus (epithelium, stroma and myometrium), or anti-Mullerian hormone type 2 receptor (Amhr2)-Cre (Tsc1(Amhr2(d/d))), which inactivates stromal and myometrial Tsc1. Tsc1(PR(d/d)) and Tsc1(Amhr2(d/d)) females are infertile resulting from oviductal hyperplasia, retention of embryos in the oviduct and implantation failure. In contrast to the appropriate embryonic development after fertilization seen in Tsc1(Amhr2(d/d)) females, embryo development was disrupted in Tsc1(PR(d/d)) females. In addition, uteri in Tsc1(PR(d/d)) and Tsc1(Amhr2(d/d)) females showed epithelial hyperplasia but not endometrial cancer. In conclusion, Tsc1(PR(d/d)) and Tsc1(Amhr2(d/d)) have overlapping yet distinct phenotypes in the context of compartment-specific deletion of Tsc1.


Assuntos
Complexos Multiproteicos/metabolismo , Oviductos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Útero/metabolismo , Animais , Estradiol/sangue , Feminino , Fertilização in vitro , Imuno-Histoquímica , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Mutantes , Complexos Multiproteicos/genética , Ovário/metabolismo , Progesterona/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina-Treonina Quinases TOR/genética , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
9.
Sci Transl Med ; 15(684): eade1857, 2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36812344

RESUMO

Obesity, defined as a body mass index (BMI) ≥ 30, is an established risk factor for breast cancer among women in the general population after menopause. Whether elevated BMI is a risk factor for women with a germline mutation in BRCA1 or BRCA2 is less clear because of inconsistent findings from epidemiological studies and a lack of mechanistic studies in this population. Here, we show that DNA damage in normal breast epithelia of women carrying a BRCA mutation is positively correlated with BMI and with biomarkers of metabolic dysfunction. In addition, RNA sequencing showed obesity-associated alterations to the breast adipose microenvironment of BRCA mutation carriers, including activation of estrogen biosynthesis, which affected neighboring breast epithelial cells. In breast tissue explants cultured from women carrying a BRCA mutation, we found that blockade of estrogen biosynthesis or estrogen receptor activity decreased DNA damage. Additional obesity-associated factors, including leptin and insulin, increased DNA damage in human BRCA heterozygous epithelial cells, and inhibiting the signaling of these factors with a leptin-neutralizing antibody or PI3K inhibitor, respectively, decreased DNA damage. Furthermore, we show that increased adiposity was associated with mammary gland DNA damage and increased penetrance of mammary tumors in Brca1+/- mice. Overall, our results provide mechanistic evidence in support of a link between elevated BMI and breast cancer development in BRCA mutation carriers. This suggests that maintaining a lower body weight or pharmacologically targeting estrogen or metabolic dysfunction may reduce the risk of breast cancer in this population.


Assuntos
Neoplasias da Mama , Glândulas Mamárias Humanas , Feminino , Humanos , Animais , Camundongos , Mutação em Linhagem Germinativa , Leptina , Glândulas Mamárias Humanas/patologia , Fosfatidilinositol 3-Quinases , Proteína BRCA2 , Proteína BRCA1/genética , Neoplasias da Mama/patologia , Dano ao DNA , Epitélio/patologia , Obesidade , Estrogênios , Mutação , Microambiente Tumoral
10.
Int J Gynecol Pathol ; 36(1): 1, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-29140255
11.
Exp Cell Res ; 317(11): 1580-9, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21397598

RESUMO

Pten is the most frequently mutated gene in uterine endometriod carcinoma (UEC) and its precursor complex atypical hyperplasia (CAH). Because the mutation frequency is similar in CAH and UEC, Pten mutations are thought to occur relatively early in endometrial tumorigenesis. Previous work from our laboratory using the Pten(+/-) mouse model has demonstrated somatic inactivation of the wild type allele of Pten in both CAH and UEC. In the present study, we injected adenoviruses expressing Cre into the uterine lumen of adult Pten floxed mice in an attempt to somatically delete both alleles of Pten specifically in the endometrium. Our results demonstrate that biallelic inactivation of Pten results in an increased incidence of carcinoma as compared to the Pten(+/-) mouse model. In addition, the carcinomas were more aggressive with extension beyond the uterus into adjacent tissues and were associated with decreased expression of nuclear ERα as compared to associated CAH. Primary cultures of epithelial and stromal cells were prepared from uteri of Pten floxed mice and Pten was deleted in vitro using Cre expressing adenovirus. Pten deletion was evident in both the epithelial and stromal cells and the treatment of the primary cultures with estrogen had different effects on Akt activation as well as Cyclin D3 expression in the two purified components. This study demonstrates that somatic biallelic inactivation of Pten in endometrial epithelium in vivo results in an increased incidence and aggressiveness of endometrial carcinoma compared to mice carrying a germline deletion of one allele and provides an important in vivo and in vitro model system for understanding the genetic underpinnings of endometrial carcinoma.


Assuntos
Neoplasias do Endométrio/patologia , Deleção de Genes , PTEN Fosfo-Hidrolase/fisiologia , Lesões Pré-Cancerosas/patologia , Adenoviridae , Animais , Western Blotting , Células Cultivadas , Neoplasias do Endométrio/metabolismo , Endométrio/citologia , Endométrio/metabolismo , Células Epiteliais/metabolismo , Feminino , Homozigoto , Técnicas Imunoenzimáticas , Integrases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Lesões Pré-Cancerosas/metabolismo , Células Estromais/metabolismo , Útero/citologia , Útero/metabolismo
12.
Gynecol Oncol ; 122(2): 424-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21570712

RESUMO

OBJECTIVE: Endometrial cancer (EMC) is the most common gynecological malignancy. The etiology and the cell types that are conducive to EMC are not completely understood, provoking further studies. Our objective was to determine whether deletion of Pten specifically in the uterine stroma and myometrium induces cancer or manifests different phenotypes. METHODS: Pten(Amhr2(d/d)) mice with conditional deletion of Pten in the mouse uterine stroma and myometrium, but not in the epithelium, were generated by mating floxed Pten mice and anti-Mullerian hormone type 2 receptor (Amhr2)-Cre mice. The phenotypes were compared between Pten(f/f) and Pten(Amhr2(d/d)) uteri. RESULTS: We show that conditional deletion of Pten in the mouse uterine stroma and myometrium, but not in the epithelium, fails to generate EMC even at the age of 5 months. Surprisingly Pten deletion by Amhr2-Cre transformed a large number of myometrial cells into adipocytes with lipid accumulation, possibly a result of increased levels of SREBP1 and PPARγ which regulate adipose differentiation. CONCLUSIONS: These results provide evidence that deletion of Pten specifically in the stroma and myometrium does not result in EMC in female mice examined up to 5 months of age but alters the myocytes to adipocytes and mimics histologic similarities with lipoleiomyomas in humans, raising the possibility of using this mouse model to further explore the cause of the disease.


Assuntos
Diferenciação Celular , Neoplasias do Endométrio/etiologia , PTEN Fosfo-Hidrolase/fisiologia , Útero/patologia , Adipócitos/citologia , Animais , Transição Epitelial-Mesenquimal , Feminino , Camundongos , Camundongos Endogâmicos BALB C , PPAR gama/genética , Fenótipo , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
13.
Gynecol Oncol ; 116(2): 286-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20109727

RESUMO

OBJECTIVE: Uterine serous carcinoma (USC) is an aggressive endometrial cancer associated with poor prognosis despite comprehensive surgical staging and adjuvant chemotherapy and radiation therapy. Biologic targets have yet to be fully explored in this disease and research on such targets could lead to clinical trials utilizing a new class of therapeutics. METHODS: A MEDLINE search of molecular alterations in USC was performed and reviewed. RESULTS: Studies evaluating primary USC tumors for molecular alterations have focused on molecules such as the transmembrane receptor ERBB2 (HER-2), the epidermal growth factor receptor (EGFR), and the recently characterized oncogene PIK3CA, which encodes the catalytic p110-alpha subunit of phosphatidylinositol 3-kinase (PI3K). In addition, claudin-3 and claudin-4 have recently been shown to be highly expressed in USC and have potential utilization as tumor markers and possible target proteins. CONCLUSIONS: Since optimal treatment of uterine serous carcinoma remains unknown, novel therapeutic approaches need to be actively pursued. The molecular targets discussed warrant further investigation and suggest a potential role for therapeutic agents targeting HER-2 and EGFR, as well as downstream targets such as the PI3K-AKT-mTOR pathway in the treatment of uterine serous carcinoma.


Assuntos
Cistadenocarcinoma Seroso/genética , Neoplasias Uterinas/genética , Animais , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Cistadenocarcinoma Seroso/terapia , Feminino , Humanos , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia , Neoplasias Uterinas/terapia
14.
Int J Gynecol Pathol ; 34(2): 111, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25675177
16.
Int J Cancer ; 124(6): 1349-57, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19101990

RESUMO

The light microscopic distinction between complex atypical hyperplasia (CAH) and invasive endometrioid carcinoma (UEC) on endometrial sampling is problematic and often has significant clinical implications. Using mouse models of endometrial tumorigenesis based on two of the most common molecular alterations found in primary human UEC we sought to characterize the transition from CAH to carcinoma to identify clinically useful biomarkers. We used the previously described Pten(+/-); Mlh1(-/-) mouse model. DNA was isolated from microdissected lesions (CAH and carcinoma) and analyzed for LOH and mutations of Pten and additional candidate genes. To identify novel candidate genes associated with invasion, global gene expression profiles were compared from uteri with extensive CAH and carcinoma. The majority of CAHs and carcinomas, arising in this model showed biallelic inactivation of Pten mediated through LOH or intragenic mutation of the wild-type allele suggesting that complete loss of Pten is insufficient for the development of carcinoma. The global gene expression studies detected increased expression of oviduct-specific glycoprotein (OGP) in carcinoma as compared with CAHs. This finding was validated using immunohistochemical staining in a collection of primary human UECs and CAHs. Our studies identify a molecular marker for invasive endometrial cancer that may have clinical significance, and highlight the usefulness of this mouse model in not only understanding the genetic underpinnings of endometrial carcinoma, but as a tool to develop clinically relevant biomarkers.


Assuntos
Neoplasias do Endométrio/patologia , Tubas Uterinas/patologia , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Biomarcadores Tumorais/análise , Análise Mutacional de DNA , Primers do DNA , Modelos Animais de Doenças , Éxons , Feminino , Genótipo , Perda de Heterozigosidade , Camundongos , Camundongos Knockout , Proteína 1 Homóloga a MutL , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética
17.
Gynecol Oncol ; 113(3): 370-3, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19272638

RESUMO

OBJECTIVES: Uterine serous carcinoma (USC) is an aggressive endometrial cancer associated with poor prognosis despite comprehensive surgical staging and adjuvant chemotherapy and radiation therapy. Biologic targets have yet to be fully explored in this disease and research on such targets could lead to clinical trials utilizing a new class of therapeutics. This study sought to evaluate primary USC tumors for molecular alterations in epidermal growth factor receptor (EGFR) and the recently characterized oncogene PIK3CA, which encodes the catalytic p110-alpha subunit of phosphatidylinositol 3-kinase (PI3K) and thus activates the AKT-mTOR oncogenic pathway. METHODS: Paraffin-embedded archival tissue of 45 primary USC tumors was utilized in this study. Immunohistochemical analysis of EGFR was performed and cases given a score of 0 to 12 calculated as the product of staining intensity (0 to 3+) and the percentage of positively stained cells (0-4), with 1=1-25%, 2=26-50%, 3=51-75%, and 4=76-100%. For mutational analysis, neoplastic tissue was microdissected and DNA was extracted with phenol-chloroform. Exons 18 through 21 of EGFR and exons 9 and 20 of PIK3CA, the most commonly mutated exons of these genes, were amplified and directly sequenced. RESULTS: When EGFR was evaluated, moderate or strong EGFR membranous staining was observed in 25/45 (56%) USC cases. Thus, a mutational analysis was performed on 35 cases, including all cases with moderate and strong EGFR staining. No mutations were identified in EGFR. In contrast, PIK3CA mutations were confirmed in 5/34 (15%) of USC cases. Four cases were mutated in exon 20 and one case was mutated in exon 9. CONCLUSIONS: Since optimal treatment of uterine serous carcinoma remains unknown, novel therapeutic approaches need to be actively pursued. In the current study of primary USC tumors, oncogenic mutations of the PIK3CA gene were seen in 15% of USC cases. This represents the first report of this gene mutation in USC. In addition, EGFR stained positively in the majority of cases, suggesting a possible target protein. These findings warrant further investigation and suggest a potential role for therapeutic agents targeting the PI3K-AKT-mTOR pathway, such as rapamycin, as well as possible targets of EGFR in the treatment of uterine serous carcinoma.


Assuntos
Carcinoma/genética , Receptores ErbB/genética , Fosfatidilinositol 3-Quinases/genética , Neoplasias Uterinas/genética , Classe I de Fosfatidilinositol 3-Quinases , Análise Mutacional de DNA , DNA de Neoplasias , Feminino , Humanos , Estudos Retrospectivos
18.
Cancer Res ; 67(9): 3998-4004, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17483309

RESUMO

Uterine cancer is the most common cancer of the female genital tract and is the fourth most frequent cause of cancer death in women in the U.S. Despite the high prevalence of uterine cancers, the molecular events that lead to neoplastic transformation in the uterus are poorly understood. Moreover, there are limited mouse models to study these malignancies. We generated transgenic mice with high-mobility group A1 gene (HMGA1a) expression targeted to uterine tissue and all female mice developed tumors by 9 months of age. Histopathologically, the tumors resemble human uterine adenosarcoma and are transplantable. To determine whether these findings are relevant to human disease, we evaluated primary human uterine neoplasms and found that HMGA1a mRNA and protein levels are increased in most high-grade neoplasms but not in normal uterine tissue, benign tumors, or most low-grade neoplasms. We also found that HMGA1a up-regulates cyclooxygenase 2 (COX-2) expression in transgenic tumors. Moreover, both HMGA1a and COX-2 expression are up-regulated in high-grade human leiomyosarcomas. Using chromatin immunoprecipitation, HMGA1a binds directly to the COX-2 promoter in human uterine cancer cells in vivo and activates its expression in transfection experiments. We also show that blocking either HMGA1a or COX-2 in high-grade human uterine cancer cells blocks anchorage-independent cell growth in methylcellulose. These findings show that HMGA1a functions as an oncogene when overexpressed in the uterus and contributes to the pathogenesis of human uterine cancer by activating COX-2 expression. Although a larger study is needed to confirm these results, HMGA1a may be a useful marker for aggressive human uterine cancers.


Assuntos
Adenossarcoma/genética , Ciclo-Oxigenase 2/biossíntese , Proteína HMGA1a/genética , Neoplasias Uterinas/genética , Adenossarcoma/enzimologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ciclo-Oxigenase 2/genética , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/biossíntese , Humanos , Infertilidade Feminina/genética , Camundongos , Camundongos Nus , Camundongos Transgênicos , Transplante de Neoplasias , Transfecção , Regulação para Cima , Neoplasias Uterinas/enzimologia
19.
Annu Rev Pathol ; 14: 339-367, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30332563

RESUMO

Endometrial cancer is the most commonly diagnosed gynecologic malignancy in the United States. Endometrioid endometrial carcinomas constitute approximately 85% of newly diagnosed cases; serous carcinomas represent approximately 3-10% of diagnoses; clear cell carcinoma accounts for <5% of diagnoses; and uterine carcinosarcomas are rare, biphasic tumors. Longstanding molecular observations implicate PTEN inactivation as a major driver of endometrioid carcinomas; TP53 inactivation as a major driver of most serous carcinomas, some high-grade endometrioid carcinomas, and many uterine carcinosarcomas; and inactivation of either gene as drivers of some clear cell carcinomas. In the past decade, targeted gene and exome sequencing have uncovered additional pathogenic aberrations in each histotype. Moreover, an integrated genomic analysis by The Cancer Genome Atlas (TCGA) resulted in the molecular classification of endometrioid and serous carcinomas into four distinct subgroups, POLE (ultramutated), microsatellite instability (hypermutated), copy number low (endometrioid), and copy number high (serous-like). In this review, we provide an overview of the major molecular features of the aforementioned histopathological subtypes and TCGA subgroups and discuss potential prognostic and therapeutic implications for endometrial carcinoma.


Assuntos
Neoplasias do Endométrio/genética , Mutação , Adenocarcinoma de Células Claras/genética , Carcinoma Endometrioide/genética , Cistadenocarcinoma Seroso/genética , DNA Polimerase II/genética , Neoplasias do Endométrio/diagnóstico , Neoplasias do Endométrio/patologia , Feminino , Humanos , Instabilidade de Microssatélites , PTEN Fosfo-Hidrolase/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Prognóstico , Proteína Supressora de Tumor p53/genética
20.
Cancer Res ; 66(7): 3375-80, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585156

RESUMO

PTEN is a tumor suppressor gene frequently mutated in human cancers. In vitro and in vivo studies have shown that PTEN can exert its tumor suppressive function through a variety of mechanisms, including regulation of cell death and cell proliferation. However, it is still unclear which of the many downstream pathways are critical in each different tissue, in vivo. Loss of PTEN is the earliest detectable genetic lesion in the estrogen-related type I (endometrioid) endometrial cancer. Pten(+/-) mice develop endometrial neoplastic lesions with full penetrance, thus providing a model system to dissect the genetic and biochemical events leading to the transition from normal to hyperplastic and neoplastic endometrial epithelium. Here, we show that loss of Pten in the mouse endometrium activates Akt and results in increased phosphorylation of estrogen receptor alpha (ERalpha) on Ser(167). ERalpha phosphorylation results, in turn, in the activation of this nuclear receptor both in vivo and in vitro, even in the absence of ligand, and in its increased ability to activate the transcription of several of its target genes. Strikingly, reduction of endometrial ERalpha levels and activity dramatically reduces the neoplastic effect of Pten loss in the endometrium, in contrast to complete estrogen depletion. Thus, we provide for the first time in vivo evidence supporting the hypothesis that loss of Pten and subsequent Akt activation result in the activation of ERalpha-dependent pathways that play a pivotal role in the neoplastic process.


Assuntos
Carcinoma Endometrioide/metabolismo , Transformação Celular Neoplásica/metabolismo , Neoplasias do Endométrio/metabolismo , Receptor alfa de Estrogênio/metabolismo , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Carcinoma Endometrioide/enzimologia , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA