Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 353
Filtrer
1.
Breast Cancer Res Treat ; 176(2): 271-289, 2019 Jul.
Article de Anglais | MEDLINE | ID: mdl-31006104

RÉSUMÉ

PURPOSE: Primary breast and prostate cancers can be cured, but metastatic disease cannot. Identifying cell factors that predict metastatic potential could guide both prognosis and treatment. METHODS: We used Cell-SELEX to screen an RNA aptamer library for differential binding to prostate cancer cell lines with high vs. low metastatic potential. Mass spectroscopy, immunoblot, and immunohistochemistry were used to identify and validate aptamer targets. Aptamer properties were tested in vitro, in xenograft models, and in clinical biopsies. Gene expression datasets were queried for target associations in cancer. RESULTS: We identified a novel aptamer (Apt63) that binds to the beta subunit of F1Fo ATP synthase (ATP5B), present on the plasma membrane of certain normal and cancer cells. Apt63 bound to plasma membranes of multiple aggressive breast and prostate cell lines, but not to normal breast and prostate epithelial cells, and weakly or not at all to non-metastasizing cancer cells; binding led to rapid cell death. A single intravenous injection of Apt63 induced rapid, tumor cell-selective binding and cytotoxicity in MDA-MB-231 xenograft tumors, associated with endonuclease G nuclear translocation and DNA fragmentation. Apt63 was not toxic to non-transformed epithelial cells in vitro or adjacent normal tissue in vivo. In breast cancer tissue arrays, plasma membrane staining with Apt63 correlated with tumor stage (p < 0.0001, n = 416) and was independent of other cancer markers. Across multiple datasets, ATP5B expression was significantly increased relative to normal tissue, and negatively correlated with metastasis-free (p = 0.0063, 0.00039, respectively) and overall (p = 0.050, 0.0198) survival. CONCLUSION: Ecto-ATP5B binding by Apt63 may disrupt an essential survival mechanism in a subset of tumors with high metastatic potential, and defines a novel category of cancers with potential vulnerability to ATP5B-targeted therapy. Apt63 is a unique tool for elucidating the function of surface ATP synthase, and potentially for predicting and treating metastatic breast and prostate cancer.


Sujet(s)
Aptamères nucléotidiques/administration et posologie , Tumeurs du sein/anatomopathologie , Membrane cellulaire/métabolisme , Mitochondrial Proton-Translocating ATPases/génétique , Mitochondrial Proton-Translocating ATPases/métabolisme , Tumeurs de la prostate/anatomopathologie , Administration par voie intraveineuse , Animaux , Aptamères nucléotidiques/pharmacologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Dépistage précoce du cancer , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Cellules MCF-7 , Mâle , Souris , Mitochondrial Proton-Translocating ATPases/antagonistes et inhibiteurs , Stadification tumorale , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Technique SELEX , Résultat thérapeutique , Régulation positive , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Breast Cancer Res Treat ; 166(1): 85-94, 2017 Nov.
Article de Anglais | MEDLINE | ID: mdl-28717852

RÉSUMÉ

PURPOSE: Elevated S100A8 expression has been observed in cancers of the bladder, esophagus, colon, ovary, and breast. S100A8 is expressed by breast cancer cells as well as by infiltrating immune and myeloid cells. Here we investigate the association of elevated S100A8 protein expression in breast cancer cells and in breast tumor stroma with survival outcomes in a cohort of breast cancer patients. PATIENTS AND METHODS: Tissue microarrays (TMA) were constructed from breast cancer specimens from 417 patients with stage I-III breast cancer treated at the University of Michigan Comprehensive Cancer Center between 2004 and 2006. Representative regions of non-necrotic tumor and distant normal tissue from each patient were used to construct the TMA. Automated quantitative immunofluorescence (AQUA) was used to measure S100A8 protein expression, and samples were scored for breast cancer cell and stromal S100A8 expression. S100A8 staining intensity was assessed as a continuous value and by exploratory dichotomous cutoffs. Associations between breast cancer cell and stromal S100A8 expression with disease-free survival and overall survival were determined using the Kaplan-Meier method and Cox proportional hazard models. RESULTS: High breast cancer cell S100A8 protein expression (as indicated by AQUA scores), as a continuous measure, was a significant prognostic factor for OS [univariable hazard ratio (HR) 1.24, 95% confidence interval (CI) 1.00-1.55, p = 0.05] in this patient cohort. Exploratory analyses identified optimal S100A8 AQUA score cutoffs within the breast cancer cell and stromal compartments that significantly separated survival curves for the complete cohort. Elevated breast cancer cell and stromal S100A8 expression, indicated by higher S100A8 AQUA scores, significantly associates with poorer breast cancer outcomes, regardless of estrogen receptor status. CONCLUSIONS: Elevated breast cancer cell and stromal S1008 protein expression are significant indicators of poorer outcomes in early stage breast cancer patients. Evaluation of S100A8 protein expression may provide additional prognostic information beyond traditional breast cancer prognostic biomarkers.


Sujet(s)
Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Calgranuline A/métabolisme , Cellules stromales/métabolisme , Marqueurs biologiques tumoraux , Tumeurs du sein/mortalité , Calgranuline A/génétique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Femelle , Technique d'immunofluorescence , Humains , Estimation de Kaplan-Meier , Grading des tumeurs , Stadification tumorale , Pronostic , Modèles des risques proportionnels , Récepteurs des oestrogènes/métabolisme , Cellules stromales/anatomopathologie , Analyse sur puce à tissus , Microenvironnement tumoral
3.
Oncogene ; 36(11): 1559-1572, 2017 03.
Article de Anglais | MEDLINE | ID: mdl-27669433

RÉSUMÉ

The receptor for advanced glycation end products (RAGE) is highly expressed in various cancers and is correlated with poorer outcome in breast and other cancers. Here we tested the role of targeting RAGE by multiple approaches in the tumor and tumor microenvironment, to inhibit the metastatic process. We first tested how RAGE impacts tumor cell-intrinsic mechanisms using either RAGE overexpression or knockdown with short hairpin RNAs (shRNAs). RAGE ectopic overexpression in breast cancer cells increased MEK-EMT (MEK-epithelial-to-mesenchymal transition) signaling, transwell invasion and soft agar colony formation, and in vivo promoted lung metastasis independent of tumor growth. RAGE knockdown with multiple independent shRNAs in breast cancer cells led to decreased transwell invasion and soft agar colony formation, without affecting proliferation. In vivo, targeting RAGE shRNA knockdown in human and mouse breast cancer cells, decreased orthotopic tumor growth, reduced tumor angiogenesis and recruitment of inflammatory cells, and markedly decreased metastasis to the lung and liver in multiple xenograft and syngeneic mouse models. To test the non-tumor cell microenvironment role of RAGE, we performed syngeneic studies with orthotopically injected breast cancer cells in wild-type and RAGE-knockout C57BL6 mice. RAGE-knockout mice displayed striking impairment of tumor cell growth compared with wild-type mice, along with decreased mitogen-activated protein kinase signaling, tumor angiogenesis and inflammatory cell recruitment. To test the combined inhibition of RAGE in both tumor cell-intrinsic and non-tumor cells of the microenvironment, we performed in vivo treatment of xenografted tumors with FPS-ZM1 (1 mg/kg, two times per week). Compared with vehicle, FPS-ZM1 inhibited primary tumor growth, inhibited tumor angiogenesis and inflammatory cell recruitment and, most importantly, prevented metastasis to the lung and liver. These data demonstrate that RAGE drives tumor progression and metastasis through distinct tumor cell-intrinsic and -extrinsic mechanisms, and may represent a novel and therapeutically viable approach for treating metastatic cancers.


Sujet(s)
Tumeurs du sein/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire , Modèles animaux de maladie humaine , Évolution de la maladie , Transition épithélio-mésenchymateuse/génétique , Femelle , Expression des gènes , Techniques de knock-down de gènes , Hétérogreffes , Humains , Ligands , Souris , Souris knockout , Mitogen-Activated Protein Kinases/métabolisme , Petit ARN interférent , Récepteur spécifique des produits finaux de glycosylation avancée/génétique , Charge tumorale
4.
Oncogene ; 34(42): 5395-405, 2015 Oct 16.
Article de Anglais | MEDLINE | ID: mdl-25659579

RÉSUMÉ

The transition from ductal carcinoma in situ (DCIS) to invasive breast cancer (IBC) is a crucial step in breast cancer progression. The specific alterations that govern this transition have not been elucidated. HER2/neu is frequently overexpressed in DCIS but is less common in IBC, thereby suggesting additional requirements for transformation. To identify genes capable of cooperating with HER2/neu to fully transform mammary epithelial cells, we used an insertional mutagenesis screen on cells isolated from wild-type neu expressing mice and identified the E3 ligase HACE1 as HER2 cooperative tumor suppressor gene. Loss of HACE1 expression is commonly seen in clinical breast cancer data sets. HACE1 downregulation in normal human mammary epithelial cells (HMECs) results in the accumulation of the activated GTP-bound Rac1 partially transforming these cells. Overexpression of HER2 activates Rac1, which further accumulates upon HACE1 loss resulting in Rac1 hyperactivation. Although the knockdown of HACE1 or overexpression of HER2 alone in HMECs is not sufficient for tumorigenesis, HER2 overexpression combined with HACE1 downregulation fully transforms HMECs resulting in robust tumor formation. The pharmaceutical interference of Rac function abrogates the effects of HACE1 loss both in vitro and in vivo, resulting in marked reduction in tumor burden. Our work supports a critical role for HACE1 in breast cancer progression and identifies patients that may benefit from Rac-targeted therapies.


Sujet(s)
Tumeurs du sein/étiologie , Transduction du signal/physiologie , Ubiquitin-protein ligases/physiologie , Protéine G rac1/physiologie , Animaux , Lignée cellulaire tumorale , Transformation cellulaire néoplasique , Évolution de la maladie , Femelle , Humains , Souris , Souris SCID , Mutagenèse par insertion , Récepteur ErbB-2/physiologie
5.
Breast Cancer Res Treat ; 143(1): 11-8, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24276281

RÉSUMÉ

CD44 is a transmembrane glycoprotein involved in numerous cellular functions, including cell adhesion and extracellular matrix interactions. It is known to be functionally diverse, with alternative splice variants increasingly implicated as a marker for tumor-initiating stem cells associated with poor prognosis. Here, we evaluate CD44 as a potential marker of long-term breast cancer outcomes. Tissue specimens from patients treated on the National Cancer Institute 79-C-0111 randomized trial of breast conservation versus mastectomy between 1979 and 1987 were collected, and immunohistochemistry was performed using the standard isoform of CD44. Specimens were correlated with patient characteristics and outcomes. Survival analysis was performed using the log rank test. Fifty-one patients had evaluable tumor sections and available long-term clinical follow up data at a median follow up of 25.7 years. Significant predictors of OS were tumor size (median OFS 25.4 years for ≤2 cm vs. 7.5 years for >2 cm, p = 0.001), nodal status (median OS 17.2 years for node-negative patients vs. 6.7 years for node positive patients, p = 0.017), and CD44 expression (median OS 18.9 years for CD44 positive patients vs. 8.6 years for CD44 negative patients, p = 0.049). There was a trend toward increased PFS for patients with CD44 positive tumors (median PFS 17.9 vs. 4.3 years, p = 0.17), but this did not reach statistical significance. These findings illustrate the potential utility of CD44 as a prognostic marker for early stage breast cancer. Subgroup analysis in patients with lymph node involvement revealed CD44 positivity to be most strongly associated with increased survival, suggesting a potential role of CD44 in decision making for axillary management. As there is increasing interest in CD44 as a therapeutic target in ongoing clinical trials, the results of this study suggest additional investigation regarding the role CD44 in breast cancer is warranted.


Sujet(s)
Tumeurs du sein/métabolisme , Tumeurs du sein/mortalité , Antigènes CD44/métabolisme , Adulte , Sujet âgé , Tumeurs du sein/anatomopathologie , Femelle , Études de suivi , Humains , Immunohistochimie , Adulte d'âge moyen , Grading des tumeurs , Métastase tumorale , Pronostic , Facteurs de risque , Charge tumorale
6.
Breast Cancer Res Treat ; 122(2): 371-80, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-19842031

RÉSUMÉ

Studies of gene regulated by estrogen in breast cancer 1 (GREB1) have focused on mRNA levels with limited evidence about GREB1 protein expression in normal and breast cancer cells. A monoclonal antibody that recognizes GREB1 protein in breast tissues could be applied to correlate protein expression with established mRNA expression data. A hybridoma expressing a murine monoclonal antibody targeting a 119 amino acid peptide specific to human GREB1 was generated. The novel monoclonal GREB1 antibody (GREB1ab) was validated for use in Western blotting as well as immunohistochemical (IHC) applications. GREB1ab detects a 216 kDa protein corresponding to GREB1 in estrogen receptor alpha (ERalpha+) breast cancer cells as well as ERalpha- breast cancer cells transduced with a GREB1 expression vector. GREB1ab specificity was verified using an ERalpha antagonist to prevent GREB1 induction as well as a silencing siRNA targeting GREB1 mRNA. GREB1ab was further validated for detection of GREB1 by IHC in breast cancer cell lines and breast tissue microarrays (TMA). ERalpha+ cell lines were observed to express GREB1 while ERalpha- cell lines did not express detectable levels of the protein. Using breast cancer tissue whole sections, IHC with the GREB1ab identified protein expression in ERalpha+ breast cancer tissue as well as normal breast tissue, with little GREB1 expression in ERalpha- breast cancer tissue. Furthermore, these data indicate that GREB1 mRNA expression correlates well with protein expression. The novel monoclonal GREB1ab is specific for GREB1 protein. This antibody will serve as a tool for investigations focused on the expression, distribution, and function of GREB1 in normal breast and breast cancer tissues.


Sujet(s)
Anticorps monoclonaux/biosynthèse , Spécificité des anticorps , Tumeurs du sein/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines tumorales/métabolisme , ARN messager/métabolisme , Animaux , Anticorps monoclonaux/génétique , Technique de Western , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Lignée cellulaire tumorale , Oestradiol/métabolisme , Antagonistes des oestrogènes/pharmacologie , Récepteur alpha des oestrogènes/antagonistes et inhibiteurs , Récepteur alpha des oestrogènes/métabolisme , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Hybridomes , Immunohistochimie , Souris , Souris de lignée BALB C , Protéines tumorales/génétique , Protéines tumorales/immunologie , Interférence par ARN , Récepteur ErbB-2/métabolisme , Reproductibilité des résultats , Analyse sur puce à tissus
7.
J Pharmacol Exp Ther ; 299(3): 849-57, 2001 Dec.
Article de Anglais | MEDLINE | ID: mdl-11714868

RÉSUMÉ

We used expression microarrays to test the effects of rifampin on the overall pattern of mRNA expression of multiple metabolic enzymes in primary human hepatocytes. Two microarrays were utilized, a cDNA-based array and one that is oligonucleotide-based. The cDNA-based expression arrays showed that rifampin caused a 7.7 +/- 6.6-fold induction in CYP2A6 and a 4.0 +/- 2.0-fold increase in the CYP2C family of enzymes while having little effect on CYP2E1 or CYP2D6. Many non-P450 enzymes were also induced including FMO-4 and -5, UGT-1A, MAO-B, and GST-P1. The oligonucleotide-based array made it possible to detect different levels of induction within the CYP2C family, with rifampin causing a 6.5-fold increase in expression of CYP2C8 and a 3.7-fold increase in CYP2C9 while having no effect on the level of CYP2C18 mRNA. Rifampin also induced other CYP enzymes including CYP2B6 and all three members of the CYP3A family, with CYP3A4 showing the highest level of induction at 55.1-fold. RNase protection assays were used to validate results from the arrays and a comparison of all three methods of mRNA detection showed qualitatively similar results. These data make it clear that rifampin treatment brings about broad changes in the pattern of gene expression, rather than increased expression of a small number of metabolic enzymes. Clinicians and researchers who use and study rifampin and other drugs that induce drug metabolism should be alert to the possibility of multiple effects.


Sujet(s)
ADN complémentaire/effets des médicaments et des substances chimiques , Hépatocytes/effets des médicaments et des substances chimiques , Rifampicine/pharmacologie , Antibiotiques antituberculeux/pharmacologie , Cellules cultivées , ADN complémentaire/métabolisme , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Hépatocytes/physiologie , Humains , Séquençage par oligonucléotides en batterie , ARN messager/effets des médicaments et des substances chimiques , ARN messager/métabolisme , Reproductibilité des résultats
8.
Cancer Res ; 61(13): 5284-8, 2001 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-11431371

RÉSUMÉ

KAI1 is a metastasis suppressor gene for human prostate cancer and is also involved in the progression of a variety of other human cancers. Previously, we have demonstrated that KAI1 expression was down-regulated in metastatic breast cancer cell lines as well as in highly aggressive breast cancer specimens. To determine whether KAI1 expression is responsible for the metastasis suppression in breast cancer, we transfected the human KAI1 cDNA into two highly malignant breast cancer cell lines, LCC6 and MDA-MB-231, which both have low levels of endogenous KAI1 expression. Parental, vector-only transfectants and KAI1 transfectant clones were injected into the mammary fat pads and tail veins, respectively, of athymic nude mice and assessed for both spontaneous and experimental lung metastasis. High KAI1 expression significantly suppressed the metastatic potential of KAI1-transfected LCC6 cells. Metastasis suppression correlated with the reduced rate of tumor growth and a decreased clonogenicity in soft agar. Furthermore, KAI1 expression significantly suppressed the in vitro cell invasion in KAI1-transfected MDA-MB-231 cells. Our results suggested that KAI1 may function as a negative regulator of breast cancer metastasis.


Sujet(s)
Antigènes CD/physiologie , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Glycoprotéines membranaires/physiologie , Protéines proto-oncogènes , Animaux , Antigènes CD/biosynthèse , Antigènes CD/génétique , Tumeurs du sein/immunologie , Adhérence cellulaire/physiologie , Division cellulaire/physiologie , ADN complémentaire/génétique , Femelle , Gènes suppresseurs de tumeur , Humains , Antigène CD82 , Tumeurs du poumon/secondaire , Glycoprotéines membranaires/biosynthèse , Glycoprotéines membranaires/génétique , Souris , Souris nude , Transfection , Cellules cancéreuses en culture
9.
J Clin Oncol ; 19(12): 3111-6, 2001 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-11408508

RÉSUMÉ

PURPOSE: To test the hypothesis that risk factors related to lifetime estrogen exposure predict breast cancer incidence and to test if any subgroups experience enhanced benefit from raloxifene. PATIENTS AND METHODS: Postmenopausal women with osteoporosis (N = 7,705), enrolled onto the Multiple Outcomes of Raloxifene Evaluation (MORE) trial, were randomly assigned to receive placebo, raloxifene 60 mg/d, or raloxifene 120 mg/d for 4 years. Breast cancer risk was analyzed by the following baseline characteristics indicative of estrogen exposure: previous hormone replacement therapy, prevalent vertebral fractures, family history of breast cancer, estradiol level, bone mineral density (BMD), body mass index, and age at menopause. Therapy-by-subgroup interactions were assessed using a logistic regression model. RESULTS: Overall, women with the highest one-third estradiol levels (> or = 12 pmol/L) had a 2.07-fold increased invasive breast cancer risk compared with women with lower levels. Raloxifene significantly reduced breast cancer risk in both the low- and high-estrogen subgroups for all risk factors examined (P <.05 for each comparison). The women with the highest BMD and those with a family history of breast cancer experienced a significantly greater therapy benefit with raloxifene, compared with the two thirds of patients with lower BMD or those without a family history, respectively; the subgroup-by-therapy interactions were significant (P =.005 and P =.015, respectively). CONCLUSION: The MORE trial confirms that increased lifetime estrogen exposure increases breast cancer risk. Raloxifene therapy reduces breast cancer risk in postmenopausal osteoporotic women regardless of lifetime estrogen exposure, but the reduction is greater in those with higher lifetime exposure to estrogen.


Sujet(s)
Tumeurs du sein/épidémiologie , Oestrogènes , Ostéoporose/traitement médicamenteux , Chlorhydrate de raloxifène/pharmacologie , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Tumeurs du sein/étiologie , Tumeurs du sein/prévention et contrôle , Méthode en double aveugle , Oestrogènes/effets indésirables , Oestrogènes/métabolisme , Oestrogènes/physiologie , Femelle , Humains , Incidence , Adulte d'âge moyen , Post-ménopause , Chlorhydrate de raloxifène/usage thérapeutique , Risque , Facteurs de risque , Modulateurs sélectifs des récepteurs des oestrogènes/usage thérapeutique , Résultat thérapeutique
10.
Breast Cancer Res Treat ; 65(2): 125-34, 2001 Jan.
Article de Anglais | MEDLINE | ID: mdl-11261828

RÉSUMÉ

Raloxifene, a selective estrogen receptor modulator approved for the prevention and treatment of postmenopausal osteoporosis, has shown a significant reduction in breast cancer incidence after 3 years in this placebo-controlled, randomized clinical trial in postmenopausal women with osteoporosis. This article includes results from an additional annual mammogram at 4 years and represents 3,004 additional patient-years of follow-up in this trial. Breast cancers were ascertained through annual screening mammograms and adjudicated by an independent oncology review board. A total of 7,705 women were enrolled in the 4-year trial; 2,576 received placebo, 2,557 raloxifene 60 mg/day, and 2,572 raloxifene 120 mg/day. Women were a mean of 66.5-years old at trial entry, 19 years postmenopause, and osteoporotic (low bone mineral density and/or prevalent vertebral fractures). As of 1 November 1999, 61 invasive breast cancers had been reported and were confirmed by the adjudication board, resulting in a 72% risk reduction with raloxifene (relative risk (RR) 0.28, 95% confidence interval (CI) 0.17, 0.46). These data indicate that 93 osteoporotic women would need to be treated with raloxifene for 4 years to prevent one case of invasive breast cancer. Raloxifene reduced the risk of estrogen receptor-positive invasive breast cancer by 84% (RR 0.16, 95% CI 0.09, 0.30). Raloxifene was generally safe and well-tolerated, however, thromboembolic disease occurred more frequently with raloxifene compared with placebo (p=0.003). We conclude that raloxifene continues to reduce the risk of breast cancer in women with osteoporosis after 4 years of treatment, through prevention of new cancers or suppression of subclinical tumors, or both. Additional randomized clinical trials continue to evaluate this effect in postmenopausal women with osteoporosis, at risk for cardiovascular disease, and at high risk for breast cancer.


Sujet(s)
Tumeurs du sein/épidémiologie , Tumeurs du sein/prévention et contrôle , Antagonistes des oestrogènes/usage thérapeutique , Chlorhydrate de raloxifène/usage thérapeutique , Modulateurs sélectifs des récepteurs des oestrogènes/usage thérapeutique , Sujet âgé , Sujet âgé de 80 ans ou plus , Australie/épidémiologie , Méthode en double aveugle , Europe/épidémiologie , Femelle , Études de suivi , Humains , Incidence , Mammographie , Adulte d'âge moyen , Nouvelle-Zélande/épidémiologie , Ostéoporose post-ménopausique/traitement médicamenteux , Post-ménopause , Facteurs de risque , Échographie mammaire , États-Unis/épidémiologie
12.
J Natl Cancer Inst ; 93(1): 51-6, 2001 Jan 03.
Article de Anglais | MEDLINE | ID: mdl-11136842

RÉSUMÉ

BACKGROUND: High mammographic density is associated with increased breast cancer risk. Previous studies have shown that estrogens increase breast density on mammograms, but the effect on mammographic density of selective estrogen receptor modulators, such as raloxifene, is unknown. We assessed changes in mammographic density among women receiving placebo, raloxifene, or conjugated equine estrogens in an osteoporosis prevention trial. METHODS: In a 5-year multicenter, double-blind, randomized, placebo-controlled osteoporosis prevention trial, healthy postmenopausal women who had undergone hysterectomy less than 15 years before the study and had no history of breast cancer received placebo, raloxifene (at one of two doses), or conjugated estrogens (ERT). Women from English-speaking investigative sites who had baseline and 2-year craniocaudal mammograms with comparable positioning (n = 168) were eligible for this analysis. Changes in mammographic density were determined by digital scanning and computer-assisted segmentation of mammograms and were analyzed with the use of analysis of variance. All statistical tests were two-sided. RESULTS: Among the four treatment groups after 2 years on study, the mean breast density (craniocaudal view) was statistically significantly greater in the ERT group than it was in the other three groups (P<0.01 for all three comparisons). Within treatment groups, the mean breast density from baseline to 2 years decreased statistically significantly in women receiving the placebo or either the higher or lower raloxifene dose (P = 0.003, P = 0.002, and P<0.001, respectively) and showed a nonstatistically significant increase in women receiving ERT. CONCLUSIONS: In an osteoporosis prevention trial, raloxifene did not increase breast density after 2 years of treatment. Raloxifene administration should not interfere with, and could even enhance, mammographic detection of new breast cancers.


Sujet(s)
Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/prévention et contrôle , Région mammaire/effets des médicaments et des substances chimiques , Région mammaire/anatomopathologie , Oestrogènes conjugués (USP)/pharmacologie , Mammographie , Chlorhydrate de raloxifène/pharmacologie , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Méthode en double aveugle , Oestrogénothérapie substitutive , Femelle , Humains , Interprétation d'images assistée par ordinateur , Mammographie/méthodes , Adulte d'âge moyen , Ostéoporose post-ménopausique/prévention et contrôle
13.
Am J Clin Pathol ; 114(4): 552-63, 2000 Oct.
Article de Anglais | MEDLINE | ID: mdl-11026101

RÉSUMÉ

Surveillance methods in Barrett esophagus (BE) using light microscopic examination of random biopsy specimens may miss focal dysplasia. In addition, dysplastic foci identified initially may not be relocated subsequently, making chemoprevention studies difficult. By using a special gastroscope, systematic mapping (4-quadrant biopsy specimens at 1-cm intervals) was performed in 22 patients (33 total mappings yielding 700 biopsy specimens). H&E, immunohistochemistry, and DNA ploidy analysis were performed. c-erbB-2 and positive Ki-67 were detected only in dysplastic sites; thus, their detection did not precede morphologically identifiable dysplasia. On the other hand, aneuploidy and p53 were detected in dysplastic and nondysplastic areas. p53 was correlated with dysplasia, and S-phase narrowly missed correlation, while aneuploidy was not correlated. PCNA and bcl-2 were ubiquitous, limiting their usefulness. On second maps, epithelial type was reidentified with 81% accuracy. A significant correlation was found between p53 and dysplasia. Sites of dysplasia and abnormal biomarkers could be relocated accurately by using endoscopic mapping. Therefore, mapping combined with biomarker studies may provide better surveillance and serve as a useful technique in chemoprevention studies.


Sujet(s)
Oesophage de Barrett/diagnostic , Marqueurs biologiques tumoraux , Endoscopie gastrointestinale , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Aneuploïdie , Oesophage de Barrett/génétique , Oesophage de Barrett/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Biopsie , ADN/analyse , Endoscopie gastrointestinale/méthodes , Femelle , Cytométrie en flux , Études de suivi , Muqueuse gastrique/métabolisme , Muqueuse gastrique/anatomopathologie , Humains , Techniques immunoenzymatiques , Mâle , Métaplasie/anatomopathologie , Adulte d'âge moyen , Reproductibilité des résultats
14.
Br J Cancer ; 83(9): 1154-60, 2000 Nov.
Article de Anglais | MEDLINE | ID: mdl-11027428

RÉSUMÉ

Angiopoietin-1 (Ang1) has been shown to act as an angiogenic promoter in embryonic angiogenesis by promoting vascular branching, pericyte recruitment and endothelial survival. We have investigated the role of Ang1 in tumour neovascularization under clinical conditions and in animal models. The expression of Ang1 in clinical breast cancer specimens was analysed by using laser-capture microdissection and reverse transcriptase-linked polymerase chain reaction (RT-PCR) on RNA isolated from the samples. Despite the expression of Ang1 in many human breast cancer cell lines, the gene was expressed in only three of 21 breast cancer clinical specimens, even though its receptor, Tie2, is abundant in the vasculature of all of these tumours. Ang1 was then overexpressed in a human breast cancer cell line (MCF-7) on its own and in conjunction with FGF1, an angiogenic factor shown to be able to increase the tumorigenicity of MCF-7 cells. High concentrations of Ang1 were produced in the conditioned media of the transfected cells (range 156-820 ng ml(-1)). However, in contrast to its physiological role as promoter of angiogenesis, overexpression of Ang1 did not enhance tumour growth, but instead caused up to a 3-fold retardation of tumour growth (P = 0.003).


Sujet(s)
Tumeurs du sein/génétique , Glycoprotéines membranaires/génétique , Angiopoïétine-1 , Animaux , Tumeurs du sein/anatomopathologie , Cellules CHO , Division cellulaire/génétique , Cricetinae , Milieux de culture conditionnés/composition chimique , Milieux de culture conditionnés/métabolisme , ADN complémentaire/génétique , Femelle , Facteur de croissance fibroblastique de type 1 , Facteur de croissance fibroblastique de type 2/génétique , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs expérimentales de la mamelle/génétique , Tumeurs expérimentales de la mamelle/anatomopathologie , Glycoprotéines membranaires/physiologie , Souris , Souris nude , ARN messager/génétique , ARN messager/métabolisme , RT-PCR , Transfection , Transplantation hétérologue , Cellules cancéreuses en culture
15.
J Natl Cancer Inst ; 92(19): 1573-81, 2000 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-11018093

RÉSUMÉ

BACKGROUND: HER2 is a membrane receptor whose overexpression is strongly associated with poor prognosis in breast carcinomas. Inhibition of HER2 activity can reduce tumor growth, which led to the development of Herceptin, an anti-HER2 monoclonal antibody (MAb) that is already in clinical use. However, the objective response rate to Herceptin monotherapy is quite low. HER2 activity can also be inhibited by the highly cytotoxic antibiotic geldanamycin (GA). However, GA is not used clinically because of its adverse toxicity. Our purpose was to enhance the inhibitory activity of anti-HER2 MAb by coupling it to GA. METHODS: We synthesized 17-(3-aminopropylamino)GA (17-APA-GA) and conjugated it to the anti-HER2 MAb e21, to form e21 : GA. The noninternalizing anti-HER2 MAb AE1 was used as a control. Internalization assays and western blot analyses were used to determine whether the anti-HER2 MAbs and their immunoconjugates were internalized into HER2-expressing cells and reduced HER2 levels. All statistical tests were two-sided. RESULTS: The immunoconjugate e21 : GA inhibited the proliferation of HER2-overexpressing cell lines better than unconjugated e21 (concentration required for 50% inhibition = 40 versus 1650 microg/mL, respectively). At 15 microg/mL, e21 : GA reduced HER2 levels by 86% within 16 hours, whereas unconjugated e21, 17-APA-GA, or AE1 : GA reduced HER2 levels by only 20%. These effects were not caused by release of 17-APA-GA from the immunoconjugate because immunoconjugates containing [(3)H]GA were stable in serum at 37 degrees C. Furthermore, e21 : GA did not significantly inhibit proliferation of the adult T-cell leukemia cell line HuT102, which is HER2 negative yet highly sensitive to GA. CONCLUSIONS: Our findings suggest that conjugating GA to internalizing MAbs enhances the inhibitory effect of the MAbs. This approach might also be applied in cellular targeting via growth factors and may be of clinical interest.


Sujet(s)
Antibiotiques antinéoplasiques/pharmacologie , Anticorps monoclonaux/pharmacologie , Tumeurs du sein/traitement médicamenteux , Immunoconjugués , Quinones/pharmacologie , Récepteur ErbB-2/métabolisme , Animaux , Antibiotiques antinéoplasiques/immunologie , Anticorps monoclonaux/usage thérapeutique , Benzoquinones , Technique de Western , Tumeurs du sein/immunologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Lactames macrocycliques , Souris , Souris de lignée BALB C , Quinones/immunologie , Récepteur ErbB-2/immunologie , Cellules cancéreuses en culture , Régulation positive
16.
J Clin Oncol ; 18(14): 2710-7, 2000 Jul.
Article de Anglais | MEDLINE | ID: mdl-10894870

RÉSUMÉ

PURPOSE: To determine the efficacy, safety, pharmacokinetics, and effect on serum angiogenic growth factors of two dose levels of thalidomide in patients with metastatic breast cancer. PATIENTS AND METHODS: Twenty-eight patients with progressive metastatic breast cancer were randomized to receive either daily 200 mg of thalidomide or 800 mg to be escalated to 1,200 mg. Fourteen heavily pretreated patients were assigned to each dose level. Each cycle consisted of 8 weeks of treatment. Pharmacokinetics and growth factor serum levels were evaluated. RESULTS: No patient had a true partial or complete response. On the 800-mg arm, 13 patients had progressive disease at or before 8 weeks of treatment and one refused to continue treatment. The dose was reduced because of somnolence to 600 mg for five patients and to 400 mg for two and was increased for one to 1,000 mg and for four to 1,200 mg. On the 200-mg arm, 12 patients had progressive disease at or before 8 weeks and two had stable disease at 8 weeks, of whom one was removed from study at week 11 because of grade 3 neuropathy and the other had progressive disease at week 16. Dose-limiting toxicities included somnolence and neuropathy. Adverse events that did not require dose or schedule modifications included constipation, fatigue, dry mouth, dizziness, nausea, anorexia, arrhythmia, headaches, skin rash, hypotension, and neutropenia. Evaluation of circulating angiogenic factors and pharmacokinetic studies failed to provide insight into the reason for the lack of efficacy. CONCLUSION: Single-agent thalidomide has little or no activity in patients with heavily pretreated breast cancer. Further studies that include different patient populations and/or combinations with other agents might be performed at the lower dose levels.


Sujet(s)
Inhibiteurs de l'angiogenèse/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Thalidomide/usage thérapeutique , Adulte , Sujet âgé , Inhibiteurs de l'angiogenèse/administration et posologie , Inhibiteurs de l'angiogenèse/pharmacocinétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Calendrier d'administration des médicaments , Facteurs de croissance endothéliale/métabolisme , Femelle , Facteur de croissance fibroblastique de type 2/métabolisme , Substances de croissance/métabolisme , Humains , Lymphokines/métabolisme , Matrix metalloproteinases/métabolisme , Adulte d'âge moyen , Métastase tumorale , Études prospectives , Thalidomide/administration et posologie , Thalidomide/pharmacocinétique , Facteur de nécrose tumorale alpha/métabolisme , Facteur de croissance endothéliale vasculaire de type A , Facteurs de croissance endothéliale vasculaire
17.
Cancer Res ; 60(11): 3081-7, 2000 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-10850460

RÉSUMÉ

Epidermal growth factor receptor vIII (EGFRvIII) is a tumor-specific, ligand-independent, constitutively active variant of the EGFR. Its expression has been detected in gliomas and various other human malignancies. To more fully characterize the function and potential biological role of EGFRvIII in regulating cell proliferation and in tumorigenesis, we transfected EGFRvIII cDNA into a nontumorigenic, interleukin 3 (IL-3)-dependent murine hematopoietic cell line (32D cells). We observed 32D cells expressing high levels of EGFRvIII (32D/EGFRvIII P5) to be capable of abrogating the IL-3-dependent pathway in the absence of ligands. In contrast, the parental cells, 32D/EGFR, 32D/ErbB-4, and 32D/ErbB-2+ErbB-3 cells, all depended on IL-3 or EGF-like ligands for growth. 32D/EGFRvIII P5 cells subjected to long-term culture conditions in the absence of IL-3 revealed further elevation of EGFRvIII expression levels. These results suggested that the IL-3-independent phenotype is mediated by EGFRvIII. The level of expression is a critical driving force for the IL-3-independent phenotype. Dose-response analysis revealed 32D/EGFRvIII cells to require 500-fold higher concentrations (50 ng/ml) of EGF to further stimulate the EGF-mediated proliferation than in the 32D/EGFR cells (100 pg/ml). Similar effects were also observed in beta-cellulin-mediated proliferation. Moreover, 32D cells expressing high levels of EGFRvIII formed large tumors in nude mice, even when no exogenous EGF ligand was administered. In contrast, no tumors grew in mice injected with 32D/EGFR, 32D/ErbB-4, and 32D/ErbB-2+ErbB-3 cells or low-expressing clone 32D/EGFRvIII C2 cells or the parental 32D cells. The changes of the ligand specificity support the notion for an altered conformation of EGFRvIII to reveal an activated ligand-independent oncoprotein with tumorigenic activity analogous to v-erbB. These studies clearly demonstrate that EGFRvIII is capable of transforming a nontumorigenic, IL-3-dependent murine hematopoietic cell line (32D cells) into an IL-3-independent and ligand-independent malignant phenotype in vitro and in vivo. To delineate the biological significance of EGFRvIII in human breast cancer, we expressed EGFRvIII in the MCF-7 human breast cancer cell line. Expression of EGFRvIII in MCF-7 cells produced a constitutively activated EGFRvIII receptor. Expression of EGFRvIII in MCF-7 cells also elevated ErbB-2 phosphorylation, presumably through heterodimerization and cross-talk. These MCF-7/EGFRvIII transfectants exhibited an approximately 3-fold increase in colony formation in 1% serum with no significant effect observed at higher percentages of serum. A similar result was also seen in anchorage-dependent assays. Furthermore, EGFRvIII expression significantly enhanced tumorigenicity of MCF-7 cells in athymic nude mice with P < 0.001. Collectively, these results provide the first evidence that EGFRvIII could play a pivotal role in human breast cancer progression.


Sujet(s)
Tumeurs du sein/métabolisme , Récepteurs ErbB/métabolisme , Animaux , Division cellulaire , ADN complémentaire/métabolisme , Récepteurs ErbB/génétique , Femelle , Cytométrie en flux , Humains , Souris , Souris nude , Transplantation tumorale , Phosphorylation , Récepteur ErbB-2/métabolisme , Facteurs temps , Transfection , Cellules cancéreuses en culture
19.
West J Med ; 172(1): 39-42, 2000 Jan.
Article de Anglais | MEDLINE | ID: mdl-18751215
20.
Microvasc Res ; 58(3): 224-37, 1999 Nov.
Article de Anglais | MEDLINE | ID: mdl-10527766

RÉSUMÉ

Angiopoietin-1 (Ang-1) and its receptor Tie-2, a trans-membrane tyrosine kinase uniquely expressed by endothelial cells, are shown by null mutation studies to be essential to developmental angiogenesis. The phenotypic abnormalities in these knockout animals suggest that Tie-2 signaling is necessary for the maintenance and expansion of the primitive capillary network. We present in vitro evidence indicating that the Ang-1/Tie-2 system participates in the regulation of capillary tubule formation and is necessary for the survival of confluent endothelial cells. Although recombinant Ang-1, which induces Tie-2 phosphorylation, has no effect on the proliferation of endothelial cells, treatment of confluent adult bovine aortic endothelial cells (ABAE) cells grown on collagen gels with Ang-1 (100 ng/ml) causes the cells to migrate into the collagen gel and form capillary-like tubules. The tubule-forming effect of Ang-1 is similar to the effect caused by FGF-2. A soluble form of the Tie-2 extracellular domain, in fivefold molar excess, blocks Ang-1-induced tubule formation. Specific elimination of Tie-2 protein expression in cultured ABAE cells as a result of transfection with an antisense oligonucleotide causes cell death in a dose-dependent manner (IC(50) = 50 nM). The antisense treatment has no effect on cells that do not express Tie-2. Cells treated with antisense oligonucleotide show a sixfold increase in the rate of apoptosis as assessed by in situ end labeling of fragmented DNA. These findings are consistent with the view that Ang-1/Tie-2 signaling is essential for both angiogenesis and endothelial cell survival.


Sujet(s)
Endothélium vasculaire/croissance et développement , Glycoprotéines membranaires/physiologie , Récepteurs à activité tyrosine kinase/physiologie , Cellules 3T3 , Angiopoïétine-1 , Animaux , Séquence nucléotidique , Vaisseaux capillaires/cytologie , Vaisseaux capillaires/croissance et développement , Vaisseaux capillaires/physiologie , Bovins , Mort cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/physiologie , Cellules cultivées , Endothélium vasculaire/cytologie , Endothélium vasculaire/physiologie , Glycoprotéines membranaires/antagonistes et inhibiteurs , Glycoprotéines membranaires/génétique , Souris , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Oligodésoxyribonucléotides antisens/pharmacologie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/génétique , Récepteur TIE-2 , Protéines recombinantes/pharmacologie , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...