Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Clin Oncol ; 29(32): 4279-85, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21990395

RESUMO

PURPOSE: HER2 (ERBB2) status is an important prognostic and predictive marker in breast carcinoma. In recent years, Genomic Health (GHI), purveyors of the Oncotype DX test, has been separately reporting HER2 by reverse transcription polymerase chain reaction (RT-PCR) to oncologists. Because of the lack of independent evaluation, this quality assurance study was undertaken to define the concordance rate between immunohistochemistry (IHC)/fluorescent in situ hybridization (FISH) and GHI RT-PCR HER2 assay. METHODS: All patients at three participating laboratories (Magee-Womens Hospital [Pittsburgh, PA], Cleveland Clinic [Cleveland, OH], and Riverside Methodist Hospital [Columbus, OH]) with available HER2 RT-PCR results from GHI were included in this study. All IHC-positive and equivocal patient cases were further evaluated and classified by FISH at respective laboratories. RESULTS: Of the total 843 patient cases, 784 (93%) were classified as negative, 36 (4%) as positive, and 23 (3%) as equivocal at the three institutions using IHC/FISH. Of the 784 negative patient cases, 779 (99%) were also classified as negative by GHI RT-PCR assay. However, all 23 equivocal patient cases were reported as negative by GHI. Of the 36 positive cases, only 10 (28%; 95% CI, 14% to 45%) were reported as positive, 12 (33%) as equivocal, and 14 (39%) as negative. CONCLUSION: There was an unacceptable false-negative rate for HER2 status with GHI HER2 assay in this independent study. This could create confusion in the decision-making process for targeted treatment and potentially lead to mismanagement of patients with breast cancer if only GHI HER2 information is used.


Assuntos
Neoplasias da Mama/química , Garantia da Qualidade dos Cuidados de Saúde , Receptor ErbB-2/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Reações Falso-Negativas , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Receptor ErbB-2/genética
3.
Breast Cancer Res Treat ; 123(3): 651-60, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19924529

RESUMO

Insulin receptor substrate-1 (IRS-1) is a cytoplasmic scaffolding protein that is phosphorylated by insulin-like growth factor-I receptor and recruits downstream effectors. Recent evidence suggests that IRS-1 has a nuclear localization and function. Here we investigated whether nuclear and cytoplasmic IRS-1 levels are associated with clinico-pathological characteristics and clinical outcome in breast cancer patients. Tissue microarrays from 1,097 patients with stage I-II breast cancer were stained by immunohistochemistry for IRS-1. Nuclear and cytoplasmic IRS-1 were scored separately according to the Allred score. Nuclear IRS-1 showed a positive association with estrogen receptor (ER) (r = 0.09, P = 0.003) and progesterone receptor (PR) (r = 0.08, P = 0.008) status and a negative correlation with lymph node involvement (r = -0.10, P = 0.001). Cytoplasmic IRS-1 did not correlate with ER or PR but showed a positive correlation with tumor size (r = 0.10, P = 0.001) and S-phase fraction (r = 0.16, P < 0.001). In univariate analysis, tamoxifen-treated patients with tumors showing positive nuclear IRS-1 had a better recurrence-free survival (RFS) (P = 0.009) and overall survival (OS) (P = 0.0007), while no association was shown between cytoplasmic IRS-1 and RFS or OS in the same group of patients. In multivariate analysis of patients receiving tamoxifen, negative nuclear IRS-1 showed a significantly reduced RFS (P = 0.046) and OS (P = 0.018). Combining both PR and nuclear IRS-1, tamoxifen-treated patients with PR+/IRS-1+ tumors had a better RFS (P = 0.0003) and OS (P < 0.0001) when compared with patients with PR-/IRS-1- tumors. In conclusion, nuclear IRS-1 may be a useful marker to predict tamoxifen response in patients with early breast cancer, particularly when assessed in combination with PR.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Biomarcadores Tumorais/análise , Neoplasias da Mama/química , Neoplasias da Mama/tratamento farmacológico , Núcleo Celular/química , Proteínas Substratos do Receptor de Insulina/análise , Tamoxifeno/uso terapêutico , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Metástase Linfática , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Receptores de Estrogênio/análise , Receptores de Progesterona/análise , Medição de Risco , Fatores de Risco , Taxa de Sobrevida , Fatores de Tempo , Análise Serial de Tecidos , Resultado do Tratamento
4.
Clin Cancer Res ; 14(2): 370-8, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18223211

RESUMO

PURPOSE: Human invasive breast cancers (IBC) show enormous histologic and biological diversity. This study comprehensively evaluated diversity in ductal carcinoma in situ (DCIS), the immediate precursors of IBCs. EXPERIMENTAL DESIGN: The extent of diversity for conventional histologic grade and standard prognostic biomarkers assessed by immunohistochemistry was evaluated in a series of pure DCIS (n = 200) compared with a contemporaneous series of IBCs (n = 200). A subset of the DCIS (n = 25) was evaluated by DNA microarrays for the presence of luminal, basal, and erbB2 intrinsic subtypes. The extent of diversity within individual cases of DCIS (n = 120) was determined by assessing multiple regions independently for histologic (nuclear) grade and several biomarkers by immunohistochemistry, which approximate microarrays in determining intrinsic subtypes. RESULTS: DCIS showed a broad distribution of conventional histologic grades and standard biomarkers ranging from well to poorly differentiated, nearly identical to IBCs. Microarrays showed the same intrinsic subtypes in DCIS as in IBCs. However, higher resolution analysis showed that multiple histologic grades, biomarker phenotypes, and intrinsic subtypes often coexist within the same DCIS, and these diverse regions probably compete for dominance. Diversity within cases of DCIS was highly correlated with mutated p53 (P = 0.0007). CONCLUSIONS: These results support the hypothesis that poorly differentiated DCIS gradually evolve from well-differentiated DCIS by randomly acquiring genetic defects resulting in increasingly abnormal cellular features. This diversity is amplified by defects resulting in genetic instability (e.g., p53 mutation), and the alterations are propagated to IBC in a manner independent of progression to invasion.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/fisiopatologia , Carcinoma Ductal de Mama/fisiopatologia , Carcinoma Intraductal não Infiltrante/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Carcinoma Intraductal não Infiltrante/classificação , Carcinoma Intraductal não Infiltrante/genética , Carcinoma Intraductal não Infiltrante/patologia , Progressão da Doença , Fator de Transcrição GATA3/metabolismo , Genes erbB-2 , Genes p53 , Humanos , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Supressora de Tumor p53/metabolismo
5.
Am J Pathol ; 171(1): 252-62, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17591970

RESUMO

Enlargement of normal terminal duct lobular units (TDLUs) by hyperplastic columnar epithelial cells is one of the most common abnormalities of growth in the adult female human breast. These hyperplastic enlarged lobular units (HELUs) are important clinically as the earliest histologically identifiable potential precursor of breast cancer. The causes of the hyperplasia are unknown but may include estrogen-simulated growth mediated by estrogen receptor-alpha, which is highly elevated in HELUs and may be fundamental to their development. The present study used DNA microarray technology and RNA from microdissected pure epithelial cells to examine changes in gene expression and molecular pathways associated with the development of HELUs from TDLUs. The results suggest that HELUs evolve from TDLUs primarily by reactivation of pathways involved in embryonic development and suppression of terminal differentiation. Changes in ERBB genes were particularly prominent, including a uniform switch in ligands for the ERBB1 receptor (14-fold decrease in epidermal growth factor and 10-fold increase in amphiregulin, respectively) in HELUs compared with TDLUs. Epidermal growth factor regulates terminal differentiation in adult breast and amphiregulin is critical to normal embryonic breast development. Because HELUs are such early potential precursors of breast cancer, targeting some of these alterations may be especially promising strategies for breast cancer prevention.


Assuntos
Neoplasias da Mama/genética , Mama/metabolismo , Mama/patologia , Hiperplasia/genética , Lesões Pré-Cancerosas/genética , Adulto , Feminino , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Transdução de Sinais
6.
Cancer Res ; 66(16): 8266-73, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16912207

RESUMO

HER-2/neu in breast cancer is associated with tamoxifen resistance, but little data exist on its interaction with estrogen deprivation or fulvestrant. Here, we used an in vivo xenograft model of estrogen receptor (ER)-positive breast cancer with HER-2/neu overexpression (MCF7/HER-2/neu-18) to investigate mechanisms of growth inhibition and treatment resistance. MCF7/HER-2/neu-18 tumors were growth inhibited by estrogen deprivation and with fulvestrant, but resistance developed in 2 to 3 months. Inhibited tumors had reductions in ER, insulin-like growth factor-I receptor (IGF-IR), phosphorylated HER-2/neu (p-HER-2/neu), and phosphorylated p42/44 mitogen-activated protein kinase (p-MAPK). p27 was increased especially in tumors sensitive to estrogen deprivation. Tumors with acquired resistance to these therapies had complete loss of ER, increased p-HER-2/neu, increased p-MAPK, and reduced p27. In contrast, IGF-IR and phosphorylated AKT (p-AKT) levels were markedly reduced in these resistant tumors. The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor gefitinib, which can block EGFR/HER-2/neu signaling, significantly delayed the emergence of resistance to both estrogen deprivation and fulvestrant. Levels of p-MAPK and p-AKT decreased with gefitinib, whereas high ER levels were restored. Eventually, however, tumors progressed in mice treated with gefitinib combined with estrogen deprivation or fulvestrant accompanied again by loss of ER and IGF-IR, increased p-HER-2/neu, high p-MAPK, and now increased p-AKT. Thus, estrogen deprivation and fulvestrant can effectively inhibit HER-2/neu-overexpressing tumors but resistance develops quickly. EGFR/HER-2/neu inhibitors can delay resistance, but reactivation of HER-2/neu and signaling through AKT leads to tumor regrowth. Combining endocrine therapy with EGFR/HER-2/neu inhibitors should be tested in clinical breast cancer, but a more complete blockade of EGFR/HER-2/neu may be optimal.


Assuntos
Neoplasias da Mama/patologia , Estrogênios/fisiologia , Receptor ErbB-2/antagonistas & inibidores , Receptores de Estrogênio/análise , Animais , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Estrogênios/deficiência , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Transplante Heterólogo
7.
Mod Pathol ; 19(2): 172-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16424892

RESUMO

Columnar cell lesions (CCLs) of the breast with low-grade/monomorphic-type cytologic atypia are being identified increasingly in biopsies performed owing to mammographic microcalcifications. The WHO Working Group on the Pathology and Genetics of Tumours of the Breast recently introduced the term 'flat epithelial atypia' (FEA) for these lesions. However, the ability of pathologists to reproducibly diagnose FEA and to distinguish it from CCLs without atypia has not been previously evaluated. Eight pathologists with an interest in breast pathology participated in a study to address this issue. The study reference pathologist provided the other seven study pathologists with a Powerpoint tutorial that included written criteria for, and representative images of, FEA and CCLs without atypia (ie, columnar cell change and columnar cell hyperplasia). Following review of the tutorial, the study pathologists examined images in Powerpoint format from 30 CCLs and were instructed to categorize each as either 'FEA' or 'not atypical'. Overall agreement among the eight pathologists was 91.8% (95% CI, 84.0-96.9%), and the multi-rater kappa value was 0.83 (95% CI, 0.67-0.94), which is within the 'excellent agreement' range. Agreement was slightly better for determining absence of FEA (92.8%: 95% CI, 84.1-97.4%), than for determining its presence (90.4%: 95% CI, 79.9-96.7%). We conclude that the diagnosis of FEA and its distinction from CCLs without atypia is highly reproducible with the use of available diagnostic criteria.


Assuntos
Neoplasias da Mama/diagnóstico , Mama/patologia , Células Epiteliais/patologia , Lesões Pré-Cancerosas/diagnóstico , Diagnóstico Diferencial , Feminino , Humanos , Variações Dependentes do Observador , Patologia Clínica/normas , Patologia Clínica/estatística & dados numéricos , Reprodutibilidade dos Testes
8.
Breast Cancer Res ; 8(1): R6, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16417654

RESUMO

INTRODUCTION: The hyperplastic enlarged lobular unit (HELU) is a common alteration in adult female human breast and is the earliest histologically identifiable lesion with premalignant potential. Growth and differentiation in normal epithelium are regulated by estrogen and progesterone, whose effects are mediated through estrogen receptor (ER)-alpha and progesterone receptor (PR). We assessed correlations between growth (proliferation and apoptosis), endogenous hormone levels (using age as a surrogate for menopausal/estrogen status), and ER-alpha/PR expression in HELUs versus adjacent normal terminal duct lobular units (TDLUs) to gain insight into potentially premalignant hyperplasia. METHODS: Proliferation (Ki67 antigen), ER-alpha, and PR were assessed by immunohistochemistry, apoptosis using the TUNEL (terminal transferase-mediated dUTP nick end-labeling) assay, and nuclear colocalization of ER-alpha and Ki67 by dual-labeled immunofluorescence in HELUs and adjacent TDLUs (n = 100-584, depending on the factor) from 324 breasts. All factors were quantified under direct microscopic visualization. ER-alpha/PR expression was semiquantified by estimating the proportion of positive cells (0 = none, 1 = or <1/100, 2 = 1/100 to 1/10, 3 = 1/10 to 1/3, 4 = 1/3 to 2/3, and 5 = or >2/3). Ki67, TUNEL, and colocalization of ER-alpha and Ki67 were scored by absolute counting (%positive). RESULTS: ER-alpha and PR expression were significantly elevated in HELUs versus adjacent TLDUs (average score: 4.5 versus 3.1 and 3.5 versus 2.1; P < 0.0001). Proliferation was also significantly higher in HELUs versus TDLUs (average 6.3% versus 2.0%; P < 0.0001). In contrast, apoptosis was significantly lower in HELUs versus TDLUs (average 0.61% versus 0.22%; P < 0.0001). Changes in proliferation and receptor expression were similar between premenopausal and postmenopausal TDLUs and HELUs, suggesting that hyperplastic cells remain responsive to regulation by estrogen. The proportion of ER-positive/proliferating cells was much higher in HELUs than TDLUs (27.6% vs. 4.9%; P < .0001). CONCLUSION: Development of HELUs is associated with increased proliferation and decreased cell death relative to normal cells. ER-alpha and PR are highly elevated in HELUs, which may contribute to the hyperplasia because they mediate hormonal regulation of growth. An understanding of the fundamental causes of increased levels of receptors and growth may lead to new strategies to prevent breast cancer.


Assuntos
Apoptose , Neoplasias da Mama/fisiopatologia , Proliferação de Células , Receptor alfa de Estrogênio/fisiologia , Estrogênios/fisiologia , Lesões Pré-Cancerosas/fisiopatologia , Receptores de Progesterona/fisiologia , Transformação Celular Neoplásica , Receptor alfa de Estrogênio/biossíntese , Feminino , Humanos , Hiperplasia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67/análise , Menopausa , Receptores de Progesterona/biossíntese
9.
Breast Cancer Res Treat ; 95(1): 7-12, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16244788

RESUMO

Nuclear metastasis-associated 1(MTA1) protein is an estrogen receptor co-repressor that regulates transcription via chromatin remodeling, and MTA1 messenger ribonucleic acid (mRNA) levels are elevated in several kinds of locally advanced and metastatic tumors relative to non-metastatic tumors. Previous studies in our laboratory mapped MTA1 into a region showing significantly lower LOH (loss of heterozygosity) in primary breast cancers with metastases compared to node-negative tumors, suggesting that epigenetic alterations of MTA1 affect metastatic potential. The present study examined immunohistochemical expression of the MTA1 protein in treated and untreated primary human breast cancers to study the relationship between MTA1 expression and clinical outcome. Node-negative tumors that overexpress MTA1 protein had recurrence risks similar to node-positive tumors. In multivariate analysis of untreated node-negative tumors, highest expression of MTA1 was associated with increased relapse risk (hazard ratio (HR)=2.72, p=0.0003 for multivariate analysis). Tamoxifen and/or anthracylcene-based chemotherapies eliminated all MTA1 associations with clinical outcome, suggesting MTA1 overexpression predicts early disease relapse, but sensitizes breast tumors to systemic therapies.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Histona Desacetilases/metabolismo , Recidiva Local de Neoplasia/metabolismo , Proteínas Repressoras/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia Combinada , Feminino , Humanos , Mastectomia , Pessoa de Meia-Idade , Invasividade Neoplásica , Recidiva Local de Neoplasia/terapia , Estadiamento de Neoplasias , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Transativadores
10.
Breast Cancer Res Treat ; 94(3): 205-11, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16267611

RESUMO

Prevention trials using incidence or mortality as endpoints require a large number of participants and long follow-up. Trials using biomarkers as endpoints would potentially require fewer participants, less time, and significantly less resources to test promising new agents for breast cancer prevention. To test this idea, a randomized trial of tamoxifen for 1 year versus observation for 1 year was conducted to determine whether tamoxifen can cause regression of hyperplastic breast tissue, whether it changes the biomarker phenotype of premalignant disease or normal breast epithelium, and if biomarkers can be used as early surrogate indicators of response to tamoxifen. Women were identified by having an abnormal mammogram and ductal hyperplasia diagnosed by core needle biopsy. Image-directed needle biopsy was repeated in the same site of the breast after 1 year. Approximately 3000 women were screened, and 265 were eligible. Sixty-three women were randomized and paired biopsies from 45 subjects were available for analysis. There was no evidence of substantial regression of hyperplasia--fewer samples showed hyperplasia at 1 year follow-up, but this was seen in both untreated and tamoxifen-treated women. There were trends for reductions in ER and PgR and trends for increases in bcl-2 in normal and hyperplastic tissue in the tamoxifen-treated arm, though these changes did not reach statistical significance. Proliferation, determined by Ki67 staining, was not significantly changed. Clinical trials of this type are difficult to carry out and modifications in trial design are needed to make this process more efficient.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Biomarcadores Tumorais/análise , Neoplasias da Mama/prevenção & controle , Tamoxifeno/uso terapêutico , Antineoplásicos Hormonais/farmacologia , Biópsia por Agulha , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Proliferação de Células , Quimioprevenção , Determinação de Ponto Final , Feminino , Humanos , Hiperplasia , Imuno-Histoquímica , Mamografia , Pessoa de Meia-Idade , Fenótipo , Tamoxifeno/farmacologia
12.
Oncogene ; 24(26): 4220-31, 2005 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-15824740

RESUMO

The majority (75%) of human breast cancers express estrogen receptor (ER). Although ER-positive tumors usually respond to antiestrogen therapies, 30% of them do not. It is not known what controls the ER status of breast cancers or their responsiveness to antihormone interventions. In this report, we document that transgenic (TG) expression of Wnt-1 in mice induces ER-positive tumors. Loss of Pten or gain of Ras mutations during the evolution of tumors in Wnt-1 TG mice has no effect on the expression of ER, but overexpression of Neu or loss of p53 leads to ER-negative tumors. Thus, our results provide compelling evidence that expression of ER in breast cancer may be influenced by specific genetic changes that promote cancer progression. These findings constitute a first step to explore the molecular mechanisms leading to ER-positive or ER-negative mammary tumors. In addition, we find that ER-positive tumors arising in Wnt-1 TG mice are refractory to both ovariectomy and the ER antagonist tamoxifen, but lose ER expression with tamoxifen, suggesting that antiestrogen selects for ER-negative tumor cells and that the ER-positive cell fraction is dispensable for growth of these tumors. This is a first report of a mouse model of antiestrogen-resistant ER-positive breast cancers, and could provide a powerful tool to study the molecular mechanisms that control antiestrogen resistance.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Neoplasias Mamárias Animais/genética , Receptores de Estrogênio/biossíntese , Animais , Antineoplásicos Hormonais/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Genes p53 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Mitógenos , Ovariectomia/veterinária , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Proteínas Tirosina Quinases , Receptor ErbB-2/genética , Receptores de Estrogênio/fisiologia , Transdução de Sinais , Tamoxifeno/farmacologia , Proteínas Supressoras de Tumor/genética , Proteínas Wnt , Proteína Wnt1
13.
Breast Cancer Res Treat ; 90(3): 249-56, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15830138

RESUMO

The predisposition of patients with lobular carcinoma in situ (LCIS) to develop invasive breast cancer (IBC) is well known. However, relatively little is known about the biologic characteristics, which may be involved in the development and progression of LCIS. This study evaluated 59 cases of LCIS (29 pure, 30 with synchronous IBC) for five biomarkers known to be important in IBC (ER, PgR, c-erbB-2, p53 and Ki-67 proliferation rate) by immunohistochemistry. A comprehensive analysis of loss-of-heterozygosity (LOH) was performed in 12 cases (10 pure, 2 with synchronous IBC) at 15 genetic loci on 9 chromosomes. LCIS demonstrated a low grade/favorable biophenotype that was not significantly different in cases with and without synchronous IBC (ER 98%, PgR 84%, c-erbB-24%, p53 19% and proliferation rate 2%). LOH was present in 80% of pure LCIS and the highest rates of LOH were at loci on 9p (30%), 16q (63%), 17p (33%) and 17q (50%). The clustering of LOH at these four foci suggests that inactivated tumor suppressor genes in these regions may be particularly important. LOH was present in both cases of LCIS with synchronous IBC and the LOH phenotype was shared by LCIS and IBC. Our findings suggest that five known prognostic factors in IBC do not have prognostic utility in LCIS. Multiple genetic mechanisms may be involved in the development of LCIS.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/genética , Carcinoma in Situ/genética , Carcinoma Lobular/genética , Invasividade Neoplásica , Neoplasias da Mama/patologia , Carcinoma in Situ/patologia , Carcinoma Lobular/patologia , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Perda de Heterozigosidade , Fenótipo
14.
J Clin Oncol ; 23(11): 2469-76, 2005 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-15753463

RESUMO

PURPOSE: To evaluate growth factor receptor cross talk with the estrogen receptor (ER) in paired clinical breast cancer specimens and in a xenograft model before tamoxifen and at tumor progression as a possible mechanism for tamoxifen resistance. METHODS: Specimen pairs from 39 patients were tissue arrayed and stained for ER, progesterone receptor (PgR), Bcl-2, c-ErbB2 (HER-2), and phosphorylated (p) p38 mitogen-activated protein kinase (MAPK), p-ERK1/2 MAPK, and p-Akt. Xenograft MCF-7 tumors before and after tamoxifen resistance were assessed for levels of p-p38. RESULTS: Pretreatment, there were strong correlations between ER, PgR, and Bcl-2, and an inverse correlation between ER and HER-2. These correlations were lost in the tamoxifen- resistant tumors and replaced by strong correlations between ER and p-p38 and p-ERK. ER expression was lost in 17% of resistant tumors. Three (11%) of the 26 tumors originally negative for HER-2 became amplified and/or overexpressed at resistance. All ER-positive tumors that overexpressed HER-2 originally or at resistance expressed high levels of p-p38. In the pretreatment and tamoxifen-resistant specimens, there were strong correlations between p-p38 and p-ERK. In the tamoxifen-resistant xenograft tumors, like the clinical samples, there was a striking increase in p-p38. CONCLUSION: The molecular pathways driving tumor growth can change as the tumor progresses. Crosstalk between ER, HER-2, p38, and ERK may contribute to tamoxifen resistance and may provide molecular targets to overcome this resistance.


Assuntos
Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/fisiopatologia , Receptor ErbB-2/fisiologia , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/fisiologia , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico , Proteínas Quinases p38 Ativadas por Mitógeno/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos Hormonais/administração & dosagem , Biópsia , Quimioterapia Adjuvante , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Transdução de Sinais , Tamoxifeno/administração & dosagem , Transplante Heterólogo
15.
Endocrinology ; 146(4): 1707-12, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15691896

RESUMO

The E6-associated protein (E6-AP) is a dual function protein. It acts as an E3 ubiquitin-protein ligase as well as a steroid hormone receptor coactivator. Considering the influence of steroid hormone receptors and their coactivators in the normal development and tumorigenesis of reproductive organs of both genders, we examined the roles of E6-AP in the tumorigenesis of breast and prostate tissues. We demonstrated that the expression of E6-AP protein is decreased in human invasive breast and prostate carcinomas compared with their adjacent normal tissues, and this down-regulation of E6-AP is accompanied by the up-regulation of estrogen receptor (ER)-alpha in breast and androgen receptor (AR) in prostate carcinomas. Furthermore, our in vivo data from E6-AP-knockout animals indicated that the expression levels of ERalpha and AR are increased in E6-AP-null mammary and prostate glands, respectively, when compared with that of normal control animals, suggesting that E6-AP modulates the protein levels of ERalpha in breast and AR in prostate glands.


Assuntos
Neoplasias da Mama/química , Neoplasias da Próstata/química , Ubiquitina-Proteína Ligases/análise , Animais , Carcinoma in Situ/química , Carcinoma Ductal de Mama/química , Receptor alfa de Estrogênio/análise , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Complexo de Endopeptidases do Proteassoma/fisiologia , Receptores Androgênicos/análise , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/fisiologia
16.
J Clin Oncol ; 23(11): 2460-8, 2005 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-15710948

RESUMO

PURPOSE: Greater understanding of the cellular response in trastuzumab-treated patients will provide insight into the clinical management of patients. PATIENTS AND METHODS: We performed a neoadjuvant trial in 35 patients with locally advanced HER-2/neu overexpressing breast cancers who received weekly trastuzumab given as a single agent for the first 3 weeks, followed by a combination of trastuzumab and docetaxel for 12 weeks before surgery. Sequential core biopsies were taken at baseline and within weeks 1 and 3 after the first dose of trastuzumab. Clinical response to trastuzumab was assessed by tumor measurements on day 22 before chemotherapy. Core biopsies were assessed by immunohistochemistry for cell cycle and proliferation (Ki67, p27, phosphorylated [p] -MAPK), apoptosis and survival (apoptotic index, p-Akt), epidermal growth factor receptor, and total and p-HER-2. RESULTS: There was early tumor regression with a median decrease of -20.0% (range. 0% to 60.4%) after only 3 weeks of trastuzumab, and eight patients (23%) had a partial response. Consistent with the clinical regressions, apoptosis was significantly induced (median increase from 3.5% to 4.7%; P = .006) within week 1, a 35% increase above baseline. No significant change in epidermal growth factor receptor score was observed in week 1, without changes in total or p-HER-2 expression. Tumors with high baseline Ki67 were less likely to respond (P = .02). CONCLUSION: In primary breast cancers, trastuzumab substantially induces apoptosis, providing a molecular explanation for both its therapeutic efficacy and its successful combination with cytotoxic chemotherapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Adulto , Idoso , Anticorpos Monoclonais Humanizados , Antígenos de Neoplasias/análise , Neoplasias da Mama/cirurgia , Ciclo Celular , Proliferação de Células , Esquema de Medicação , Feminino , Humanos , Infusões Intravenosas , Pessoa de Meia-Idade , Terapia Neoadjuvante , Estudos Prospectivos , Receptor ErbB-2 , Trastuzumab
17.
Cancer Epidemiol Biomarkers Prev ; 14(1): 41-7, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15668474

RESUMO

We evaluated associations between reproductive and lifestyle risk factors with breast cancer tumor marker status in a case-control study. Cases were premenopausal women living in Vietnam and China who were eligible for a clinical trial of oophorectomy and tamoxifen as treatment for breast cancer (n = 682). Controls were nonrelative hospital visitors, matched on age to the cases (n = 649). Immunohistochemical analysis was used to identify the presence of estrogen receptor (ER) and progesterone receptor and the overexpression of HER-2/neu oncogene. Odds ratios (OR) and 95% confidence intervals (95% CI) were estimated using unconditional logistic regression, adjusted for known confounders. Overall, 280 (61%) tumor samples were ER positive and 176 (38%) were ER negative. HER-2/neu overexpression was detected in 161 (35%) samples, whereas 286 (26%) samples were HER-2/neu negative. We observed an inverse trend between increasing parity and decreasing breast cancer risk (P = 0.002). Women ages > or =25 years at first birth had increased breast cancer risk compared with women ages <25 years at first birth (OR, 1.53; 95% CI, 1.20-1.95). Women who consumed alcohol had increased risk of breast cancer compared with women who did not (OR,1.85; 95% CI, 1.32-2.61). Compared with controls, OR estimates for breast cancer by parity and age at first birth were significantly associated with ER and/or HER-2/neu tumor status by Wald test (P < 0.05). Family history, age at menarche, cumulative lactation, body mass index, and education were not significantly related to breast cancer risk. Our findings support the hypothesis that some breast cancer risk factors differ by ER and HER-2/neu tumor marker subtypes.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/etiologia , Adulto , Consumo de Bebidas Alcoólicas/epidemiologia , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/terapia , Estudos de Casos e Controles , China/epidemiologia , Terapia Combinada , Feminino , Genes erbB-2 , Humanos , Entrevistas como Assunto , Estilo de Vida , Modelos Logísticos , Idade Materna , Pessoa de Meia-Idade , Ovariectomia , Paridade , Pré-Menopausa , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Fatores de Risco , Tamoxifeno/uso terapêutico , Vietnã/epidemiologia
18.
Mod Pathol ; 17(12): 1545-54, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15272277

RESUMO

Progesterone receptor is a surrogate marker of estrogen receptor activity in breast cancer and its utility in helping predict clinical outcome has been established using biochemical assays. However, most laboratories worldwide have switched to immunohistochemistry to assess progesterone receptor, but unfortunately no validated immunohistochemical assay exists for progesterone receptor. The purpose of this study was to develop and validate an immunohistochemical assay for progesterone receptor in breast cancer. The assay was based on monoclonal antibody 1294 (DakoCytomation) and slides were scored microscopically using the 'Allred score' on a scale of 0-8. The assay was compared to ligand-binding assay in 1235 breast cancers, and a subset (n=362) that received only hormonal therapy was used to define a cutoff for progesterone receptor-positive. Clinical utility was validated in an independent set of samples (n=423) from a clinical trial randomizing premenopausal breast cancer patients to tamoxifen+oophorectomy vs observation following surgery. A cutoff of >2 (corresponding to >1% positive cells) dichotomized patients with significantly better or worse clinical outcome (P=0.0014). Progesterone receptor by immunohistochemistry provided significantly better results than progesterone receptor by ligand-binding assay in predicting clinical outcome. In the clinical trial, a positive result in univariate analyses was associated with significantly improved disease-free and overall survival both in untreated (hazard ratios/P=0.656/0.060 and 0.479/0.005, respectively) and hormonally treated patients (hazard ratios/P=0.529/0.017 and 0.451/0.007, respectively). Positive progesterone receptor remained significant for improved disease-free and overall survival (hazard ratios/P=0.666/0.038 and 0.549/0.007, respectively) in multivariate analyses including the standard variables of tumor size, nodal status, treatment, histological grade, and HER-2/neu status. Estrogen and progesterone receptors are codependent variables and progesterone receptor was a weaker predictor of response to endocrine therapy than estrogen receptor when both were included in multivariate analysis. This is the first comprehensive study assessing the clinical usefulness of progesterone receptor by immunohistochemistry in archival tissue in breast cancer. Progesterone receptor assessed by immunohistochemistry provides useful information about clinical outcome and it is better than progesterone receptor measured by ligand-binding assay.


Assuntos
Neoplasias da Mama/patologia , Imuno-Histoquímica/métodos , Receptores de Progesterona/análise , Ligação Competitiva , Neoplasias da Mama/metabolismo , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica/normas , Ligantes , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Receptores de Progesterona/metabolismo , Análise de Sobrevida
19.
Clin Cancer Res ; 10(1 Pt 2): 331S-6S, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14734488

RESUMO

Introduced more than 100 years ago, endocrine therapy is still the most important systemic therapy for all stages of estrogen receptor (ER) -positive breast tumors. A major clinical problem limiting the usefulness of this therapy is tumor resistance, either de novo or acquired during the course of the treatment. Relatively new discoveries emphasize the complexity of ER signaling and its multiple regulatory interactions with growth factor and other kinase signaling pathways. Both genomic (nuclear) and nongenomic (membrane and cytoplasmic) ER activities contribute to this intimate cross-talk, which is probably a fundamental factor in endocrine resistance. New targeted therapies, especially against the epidermal growth factor receptor/HER-2 pathway, should be carefully evaluated in more (bio)logical strategies to enable them to be exploited appropriately. A strategy of combining endocrine therapy (particularly tamoxifen) with these inhibitors, to circumvent de novo and acquired resistance, will be discussed. We will also emphasize open questions and future challenges in the dynamic research field of molecular ER biology from the endocrine therapy perspective.


Assuntos
Substâncias de Crescimento/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/metabolismo , Receptores ErbB/metabolismo , Humanos , Modelos Biológicos , Ligação Proteica , Receptor Cross-Talk , Transdução de Sinais
20.
J Natl Cancer Inst ; 95(24): 1825-33, 2003 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-14679152

RESUMO

BACKGROUND: Although antiestrogens have been effective in preventing estrogen receptor (ER)-positive breast cancer, chemopreventive agents are still needed to prevent ER-negative breast cancer. Tyrosine kinase inhibitors are promising agents for the treatment and prevention of human cancers. ZD1839 (gefitinib or Iressa) is an orally active epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor that blocks signal transduction pathways in epithelial cells. We examined whether ZD1839 blocks signal transduction and prevents the development of ER-negative breast cancer. METHODS: The ability of ZD1839 to block signal transduction in normal, immortalized, and malignant breast cells was assessed by western blotting with specific antibodies to detect phosphorylation of cytoplasmic signaling molecules. The effect of ZD1839 on growth of these breast cells was assessed with anchorage-dependent and anchorage-independent growth assays. Its effect on ER-negative mammary tumorigenesis was assessed in MMTV-erbB2 transgenic mice. All statistical tests were two-sided. RESULTS: ZD1839 suppressed the phosphorylation of EGFR and mitogen-activated protein kinase in normal and malignant breast cells. ZD1839 treatment statistically significantly suppressed mammary tumorigenesis in MMTV-erbB2 transgenic mice; median time to tumor development was approximately 230 days in vehicle-treated mice and more than 310 days in mice treated with ZD1839 at 100 mg/kg (P<.001). ZD1839 reduced proliferation of normal breast cells by 20.3% (95% confidence interval [CI] = -13.7% to 44.2%) and of tumor cells by 42.0% (95% CI = 20.2% to 58.2%). ZD1839 also increased expression of the cell cycle regulator p27 in normal mammary tissue by 48.7% (95% CI = 27.0% to 74.2%) and in tumor tissue by 50.3% (95% CI = 35.8% to 66.7%). CONCLUSION: This study appears to provide the preclinical rationale for the development of these EGFR tyrosine kinase inhibitors for the prevention of human breast cancer.


Assuntos
Anticarcinógenos/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/prevenção & controle , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Musculares , Quinazolinas/farmacologia , Receptores de Estrogênio/metabolismo , Animais , Western Blotting , Neoplasias da Mama/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células Epiteliais , Receptores ErbB/metabolismo , Feminino , Gefitinibe , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes erbB-2/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/análise , Fosforilação/efeitos dos fármacos , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/prevenção & controle , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...