Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 41
Filtrer
2.
Ann Oncol ; 33(3): 321-329, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34954044

RÉSUMÉ

BACKGROUND: In the primary analysis of the HER2CLIMB trial, tucatinib added to trastuzumab and capecitabine significantly improved overall survival (OS) and progression-free survival (PFS) in patients with human epidermal growth factor receptor 2 positive (HER2+) metastatic breast cancer. We report efficacy and safety outcomes, including the final OS and safety outcomes from follow-up in HER2CLIMB. PATIENTS AND METHODS: HER2CLIMB is a randomized, double-blind, placebo-controlled trial in patients with locally advanced or metastatic HER2+ breast cancer, including patients with brain metastases. Patients were randomized 2 : 1 to receive tucatinib or placebo, in combination with trastuzumab and capecitabine. After the primary analysis (median follow-up of 14 months), the protocol was amended to allow for unblinding sites to treatment assignment and cross-over from the placebo combination to the tucatinib combination. Protocol prespecified descriptive analyses of OS, PFS (by investigator assessment), and safety were carried out at ∼2 years from the last patient randomized. RESULTS: Six hundred and twelve patients enrolled in the HER2CLIMB trial. At a median OS follow-up of 29.6 months, median duration of OS was 24.7 months for the tucatinib combination group versus 19.2 months for the placebo combination group [hazard ratio (HR) for death: 0.73, 95% confidence interval (CI): 0.59-0.90, P = 0.004] and OS at 2 years was 51% and 40%, respectively. HRs for OS across prespecified subgroups were consistent with the HR for the overall study population. Median duration of PFS was 7.6 months for the tucatinib combination group versus 4.9 months for the placebo combination group (HR for progression or death: 0.57, 95% CI: 0.47-0.70, P < 0.00001) and PFS at 1 year was 29% and 14%, respectively. The tucatinib combination was well tolerated with a low rate of discontinuation due to adverse events. CONCLUSIONS: With additional follow-up, the tucatinib combination provided a clinically meaningful survival benefit for patients with HER2+ metastatic breast cancer.


Sujet(s)
Tumeurs du cerveau , Tumeurs du sein , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/secondaire , Tumeurs du sein/anatomopathologie , Capécitabine , Survie sans rechute , Femelle , Humains , Oxazoles , Pyridines , Quinazolines , Récepteur ErbB-2/métabolisme , Analyse de survie , Trastuzumab
3.
Oncogene ; 32(19): 2463-74, 2013 May 09.
Article de Anglais | MEDLINE | ID: mdl-22751112

RÉSUMÉ

Truncated-ERBB2 isoforms (t-ERBB2s), resulting from receptor proteolysis or alternative translation of the ERBB2 mRNA, exist in a subset of human breast tumors. t-ERBB2s lack the receptor extracellular domain targeted by therapeutic anti-ERBB2 antibodies and antibody-drug conjugates, including trastuzumab, trastuzumab-DM1 and pertuzumab. In clinical studies, expression of t-ERBB2 in breast tumors correlates with metastasis as well as trastuzumab resistance. By using a novel immuno-microarray method, we detect a significant t-ERBB2 fraction in 18 of 31 (58%) of immunohistochemistry (IHC)3+ ERBB2+ human tumor specimens, and further show that t-ERBB2 isoforms are phosphorylated in a subset of IHC3+ samples (10 of 31, 32%). We investigated t-ERBB2 biological activity via engineered expression of full-length and truncated ERBB2 isoforms in human mammary epithelial cells (HMECs), including HMEC and MCF10A cells. Expression of p110 t-ERBB2, but not p95m (m=membrane, also 648CTF) or intracellular ERBB2s, significantly enhanced cell migration and invasion in multiple cell types. In addition, only expression of the p110 isoform led to human breast epithelial cell (HMLE) xenograft formation in vivo. Expression of t-ERBB2s did not result in hyperactivation of the phosphoinositide kinase-3/AKT or mitogen-activated protein kinase signaling pathways in these cells; rather, phosphoproteomic array profiling revealed attenuation of phosphorylated signal transducer and activator of transcription 5 (STAT5) in p110-t-ERBB2-expressing cells compared to controls. Short hairpin-mediated silencing of STAT5 phenocopied p110-t-ERBB2-driven cell migration and invasion, while expression of constitutively active STAT5 reversed these effects. Thus, we provide novel evidence that (1) expression of p110 t-ERBB2 is sufficient for full transformation of HMEC, yielding in vivo xenograft formation, and (2) truncated p110 t-ERBB2 expression is associated with decreased phosphorylation of STAT5.


Sujet(s)
Tumeurs du sein/anatomopathologie , Mouvement cellulaire/physiologie , Récepteur ErbB-2/métabolisme , Facteur de transcription STAT-5/métabolisme , Animaux , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie , Femelle , Humains , Immunohistochimie , Souris , Souris de lignée NOD , Souris SCID , Invasion tumorale , Fragments peptidiques/métabolisme , Phosphorylation , Facteur de transcription STAT-5/génétique , Transduction du signal , Transplantation hétérologue
4.
Clin Breast Cancer ; 12(1): 10-8, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22154153

RÉSUMÉ

Trastuzumab improves response rate, time to progression, and overall survival when combined with first-line chemotherapy in patients with human epidermal growth factor receptor 2-positive (HER2-positive) metastatic breast cancer (MBC). However, the benefits of continuing trastuzumab beyond disease progression have not been clearly established. The literature was reviewed to obtain data on trastuzumab use beyond disease progression. In general, data from retrospective and observational studies suggest that there may be clinical benefit when trastuzumab is used beyond disease progression. These results are supported by prospective non-randomized studies. Response rates and survival outcomes have generally been superior in patients who have continued trastuzumab after disease progression compared with those who have not. Moreover, recent data from two prospective randomized phase III trials have shown that adding trastuzumab to the treatment regimen in patients with MBC who have progressed on trastuzumab-based therapy significantly prolongs progression-free survival. Emerging evidence from randomized controlled trials supports the potential clinical utility of continuing trastuzumab-based therapy beyond progression and supports the National Comprehensive Cancer Network recommendation to consider this treatment approach. Future treatment of HER2-positive MBC may involve trastuzumab being used in successive regimens in combination with other targeted therapies.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Évolution de la maladie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Essais cliniques de phase III comme sujet , Médecine factuelle , Femelle , Prévision , Humains , Études prospectives , Essais contrôlés randomisés comme sujet , Récepteur ErbB-2/métabolisme , Études rétrospectives , Trastuzumab , Résultat thérapeutique
5.
Breast Cancer Res Treat ; 132(1): 215-23, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22138748

RÉSUMÉ

NOV-002 (a formulation of disodium glutathione disulfide) modulates signaling pathways involved in tumor cell proliferation and metastasis and enhances anti-tumor immune responsiveness in tumor models. The addition of NOV-002 to chemotherapy has been shown to increase anti-tumor efficacy in animal models and some early phase oncology trials. We evaluated the clinical effects of NOV-002 in primary breast cancer, whether adding NOV-002 to standard preoperative chemotherapy increased pathologic complete response rates (pCR) at surgery, and determined whether NOV-002 mitigated hematologic toxicities of chemotherapy and whether levels of myeloid derived suppressor cells (MDSC) were predictive of response. Forty-one women with newly diagnosed stages II-IIIc HER-2 negative breast cancer received doxorubicin-cyclophosphamide followed by docetaxel (AC â†’ T) every 3 weeks and concurrent daily NOV-002 injections. The trial was powered to detect a doubling of pCR rate from 16 to 32% with NOV-002 plus AC â†’ T (α = 0.05, ß = 80%). Weekly complete blood counts were obtained as well as circulating MDSC levels on day 1 of each cycle were quantified. Of 39 patients with 40 evaluable tumors, 15 achieved a pCR (38%), meeting the primary endpoint of the trial. Concurrent NOV-002 resulted in pCR rates for AC â†’ T chemotherapy higher than previously reported. Patients with lower levels of circulating MDSCs at baseline and on the last cycle of chemotherapy had significantly higher probability of a pCR (P = 0.02). Further evaluation of NOV-002 in a randomized study is warranted.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Récepteur ErbB-2/métabolisme , Adolescent , Adulte , Sujet âgé , Antinéoplasiques/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/chirurgie , Cisplatine/administration et posologie , Association thérapeutique , Cyclophosphamide/administration et posologie , Survie sans rechute , Docetaxel , Doxorubicine/administration et posologie , Association médicamenteuse , Femelle , Disulfure de glutathion/administration et posologie , Humains , Immunité cellulaire/effets des médicaments et des substances chimiques , Estimation de Kaplan-Meier , Mastectomie , Adulte d'âge moyen , Traitement néoadjuvant , Invasion tumorale , Stadification tumorale , Taxoïdes/administration et posologie , Résultat thérapeutique , Jeune adulte
6.
Br J Cancer ; 102(5): 827-32, 2010 Mar 02.
Article de Anglais | MEDLINE | ID: mdl-20160731

RÉSUMÉ

BACKGROUND: HuHMFG1 (AS1402) is a humanised monoclonal antibody that has undergone a phase I trial in metastatic breast cancer. The aim of this study was to characterise the pharmacokinetics (PKs) of HuHMFG1 using a population PK model. METHOD: Data were derived from a phase I study of 26 patients receiving HuHMFG1 at doses ranging from 1 to 16 mg kg(-1). Data were analysed using NONMEM software and covariates were included. A limited sampling strategy (LSS) was developed using training and a validation data set. RESULTS: A linear two-compartment model was shown to be adequate to describe data. Covariate analysis indicated that weight was not related to clearance. An LSS was successfully developed on the basis of the model, in which one sample is collected immediately before the start of an infusion and the second is taken at the end of infusion. CONCLUSION: A two-compartment population PK model successfully describes HuHMFG1 behaviour. The model suggests using a fixed dose of HuHMFG1, which would simplify dosing. The model could be used to optimise dose level and dosing schedule if more data on the correlation between exposure and efficacy become available from future studies. The derived LSS could optimise further PK assessment of this antibody.


Sujet(s)
Anticorps monoclonaux/pharmacocinétique , Tumeurs du sein/métabolisme , Glycolipides/immunologie , Glycoprotéines/immunologie , Adulte , Sujet âgé , Anticorps monoclonaux humanisés , Essais cliniques de phase I comme sujet , Femelle , Humains , Gouttelettes lipidiques , Adulte d'âge moyen , Modèles biologiques , Pronostic , Études par échantillonnage , Taux de survie , Résultat thérapeutique
7.
Ann Oncol ; 20(6): 1026-31, 2009 Jun.
Article de Anglais | MEDLINE | ID: mdl-19179558

RÉSUMÉ

BACKGROUND: This phase II study evaluated the efficacy and safety of lapatinib in patients with human epidermal growth factor receptor 2 (HER2)-positive advanced or metastatic breast cancer that progressed during prior trastuzumab therapy. PATIENTS AND METHODS: Women with stage IIIB/IV HER2-overexpressing breast cancer were treated with single-agent lapatinib 1250 or 1500 mg once daily after protocol amendment. Tumor response according to RECIST was assessed every 8 weeks. HER2 expression was assessed in tumor tissue by immunohistochemistry and FISH. RESULTS: Seventy-eight patients were enrolled in the study. Investigator and independent review response rates [complete response (CR) or partial response (PR)] were 7.7% and 5.1%, and clinical benefit rates (CR, PR, or stable disease for >or=24 weeks) were 14.1% and 9.0%, respectively. Median time to progression was 15.3 weeks by independent review, and median overall survival was 79 weeks. The most common treatment-related adverse events were rash (47%), diarrhea (46%), nausea (31%), and fatigue (18%). CONCLUSIONS: Single-agent lapatinib has clinical activity with manageable toxic effects in HER2-overexpressing breast cancer that progressed on trastuzumab-containing therapy. Studies of lapatinib-based combination regimens with chemotherapy and other targeted therapies in metastatic and earlier stages of breast cancer are warranted.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Quinazolines/usage thérapeutique , Adulte , Sujet âgé , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux humanisés , Tumeurs du sein/anatomopathologie , Tumeurs du sein/secondaire , Évolution de la maladie , Femelle , Humains , Lapatinib , Adulte d'âge moyen , Récepteur ErbB-2/biosynthèse , Trastuzumab , Échec thérapeutique , Résultat thérapeutique
8.
Ann Oncol ; 17(4): 597-604, 2006 Apr.
Article de Anglais | MEDLINE | ID: mdl-16403812

RÉSUMÉ

BACKGROUND: Survivin, a novel inhibitor of apoptosis, is one of the most cancer-specific proteins identified to date. In this study we (a) evaluated the association between survivin and HER2, vascular endothelial growth factor (VEGF) and uPA/PAI-1 expression and (b) defined its effect on clinical outcome in a large breast cancer patient cohort. PATIENTS AND METHODS: Survivin expression was measured by ELISA in primary breast cancer tissue extracts from 420 patients with long-term clinical follow-up. RESULTS: Survivin was detected in 378 (90%) of the 420 primary breast cancer cases. Increased survivin levels were significantly associated with high nuclear grade (P < 0.0001), negative hormone receptor status (P = 0.0028), HER2 overexpression (P = 0.0094), VEGF expression (P < 0.0001), high uPA (P = 0.0002) and PAI-1 levels (P = 0.0002). Using the 25th percentile (1.4 ng/mg) as a cut-off point, patients expressing elevated survivin had a significantly worse disease-free survival (DFS: P = 0.0007, RR 1.97) and overall survival (OS: P = 0.0009, RR 2.11) compared with patients expressing lower levels of survivin. In multivariate analysis, this prognostic value of survivin was independent of both traditional and novel clinicopathologic factors for both DFS (P = 0.0076, RR 1.72) and OS (P = 0.0155, RR 1.76). CONCLUSIONS: The independent prognostic relevance of survivin, when combined with previous data from model systems implicating survivin in the inhibition of apoptosis, suggests that survivin may be a suitable target for future therapeutic strategies.


Sujet(s)
Tumeurs du sein/métabolisme , Protéines associées aux microtubules/métabolisme , Protéines tumorales/métabolisme , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Récepteur ErbB-2/métabolisme , Résultat thérapeutique , Activateur du plasminogène de type urokinase/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Études de cohortes , Femelle , Humains , Protéines IAP , Survivine
9.
Tech Coloproctol ; 8 Suppl 1: s50-2, 2004 Nov.
Article de Anglais | MEDLINE | ID: mdl-15655642

RÉSUMÉ

BACKGROUND: To evaluate the activity and safety of bevacizumab when given with standard 5FU/leukovorin (LV) regimens in patients with metastatic colorectal cancer who have failed irinotecan and oxaliplatin-based treatments. METHODS: Bevacizumab was given at 5 mg/kg as an IV infusion every 2 weeks. Patients received 5FU according to Roswell Park or the de Gramont regimen. RESULTS: Nineteen patients enrolled, median age 60, median PS: 1. Most common toxicity attributable to bevacizumab was mild hypertension, epistaxis and mild proteinuria; 1 patient had a CNS haemorrhage. The median number of cycles was 1 (8 weeks). Clinical benefit as disease stabilisation lasting 2-6 months was noted in 9 patients, whereas 10 progressed (median f/u: 5 months). TTP was 16 weeks, and the overall survival has not been reached (24+ weeks). CONCLUSIONS: Bevacizumab may result in growth arrest and clinical benefit in a substantial proportion of patients with colorectal cancer and no alternative treatment.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Fluorouracil/administration et posologie , Leucovorine/administration et posologie , Thérapie de rattrapage , Sujet âgé , Anticorps monoclonaux humanisés , Bévacizumab , Tumeurs colorectales/mortalité , Intervalles de confiance , Relation dose-effet des médicaments , Calendrier d'administration des médicaments , Association de médicaments , Femelle , Études de suivi , Humains , Perfusions veineuses , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Stadification tumorale , Appréciation des risques , Méthode en simple aveugle , Analyse de survie , Résultat thérapeutique
10.
Breast Cancer Res Treat ; 67(3): 223-33, 2001 Jun.
Article de Anglais | MEDLINE | ID: mdl-11561768

RÉSUMÉ

The purpose of this study was to analyze the drug interactions of paclitaxel (PTX) with epirubicin (EPI), carboplatin (CBDCA), gemcitabine (GEM) and vinorelbine (VIN) in human breast cancer cells and compare the cytotoxic activity of each drug combination in primary breast cancer samples. These experiments were intended to identify the most active agents in combination with PTX, and to provide a preclinical rational for future clinical investigations in breast cancer. Multiple drug effect/combination index (CI) isobologram analysis was applied to combinations of PTX with either CBDCA, EPI, GEM or VIN in MCF-7, MDA-MB-231 and SK-BR-3 human breast cancer cell lines. Drug concentrations were limited to the ranges achievable in humans in vivo, and the drugs were applied simultaneously at fixed molar ratios for each drug combination. Interactions were assessed at multiple effect levels (IC10-IC90). Additionally, the cytotoxic activity of these combinations was assessed in tumor samples of 50 primary breast cancer patients, utilizing the ATP-tumorchemosensitivity assay (ATP-TCA). Drug interactions were shown to be strongly dose-related in the human breast cancer cell lines investigated. At clinically relevant concentrations, CBDCA/PTX demonstrated synergistic (MCF-7) or additive (MDA-MB-231, SK-BR-3) interactions, and EPI/PTX showed additive (SK-BR-3, MCF-7) and antagonistic (MDA-MB-231) interactions. GEM/PTX and VIN/PTX, however, demonstrated antagonism over multiple dose effect levels at clinically relevant drug concentrations in all three cell lines tested. At plasma peak concentrations, EPI/PTX, CBDCA/PTX, GEM/PTX and VIN/PTX achieved > or = 90% tumor growth inhibition in 93, 86, 63 and 50%, respectively, of primary breast cancer samples investigated with the ATP-TCA. Cumulative dose-response plots of primary breast cancer tumor cells responding in vitro with > or = 90% growth inhibition showed a strong dose dependence for both EPI/PTX and CBDCA/PTX. In conclusion, the current data indicate favorable drug interactions for CBDCA/PTX at clinically relevant drug concentrations in breast cancer cells, and demonstrate superior in vitro cytotoxicity of EPI/PTX and CBDCA/PTX compared to GEM/PTX and VIN/PTX in primary breast cancer cultures.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Tumeurs du sein/anatomopathologie , Désoxycytidine/analogues et dérivés , Paclitaxel/pharmacologie , Vinblastine/analogues et dérivés , Antinéoplasiques/pharmacocinétique , Antinéoplasiques d'origine végétale/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/pharmacocinétique , Carboplatine/pharmacocinétique , Carboplatine/pharmacologie , Désoxycytidine/pharmacocinétique , Désoxycytidine/pharmacologie , Relation dose-effet des médicaments , Interactions médicamenteuses , Tests de criblage d'agents antitumoraux , Épirubicine/pharmacocinétique , Épirubicine/pharmacologie , Femelle , Humains , Paclitaxel/pharmacocinétique , Cellules cancéreuses en culture , Vinblastine/pharmacocinétique , Vinblastine/pharmacologie , Vinorelbine ,
11.
Clin Cancer Res ; 7(8): 2448-57, 2001 Aug.
Article de Anglais | MEDLINE | ID: mdl-11489825

RÉSUMÉ

PURPOSE: Recent studies suggest that HER-2/neu specifically promotes the invasive capacity of tumor cells by up-regulating secretion of the proteolytic enzyme, urokinase-type plasminogen activator (uPA), or its inhibitor, plasminogen activator inhibitor-1 (PAI-1), in colon and gastric cancer. It was the purpose of this study to: (a) evaluate the association between HER-2/neu and uPA and PAI-1 expression in a large primary breast cancer cohort; (b) perform the first multivariate analysis, including HER-2/neu, uPA, and PAI-1 in breast cancer; and (c) define the effect of HER-2/neu overexpression on uPA and PAI-1 expression in breast cancer cells. EXPERIMENTAL DESIGN: HER-2/neu, uPA, and PAI-1 were measured as continuous variables by ELISA in primary breast cancer tissue extracts from 587 patients with clinical follow-up and analyzed for correlations with clinical outcome. Furthermore, a full-length human HER-2/neu cDNA was introduced into five human breast cancer cell lines to define the effects of HER-2/neu overexpression on uPA and PAI-1 expression. In addition, we tested whether HER-2/neu antibodies could reverse any given alteration of uPA and PAI-1 levels. RESULTS: Our findings indicate a weak positive association between HER-2/neu and uPA (r = 0.147; P < 0.001) and no association between HER-2/neu and PAI-1 (r = 0.07; P = 0.085). HER-2/neu overexpression (> or =400 fmol/mg) and high levels of uPA/PAI-1 (> or =5.5 ng/mg and/or > or =14 ng/mg, respectively) were significantly associated with shorter disease-free survival (DFS; P < 0.001 and P = 0.003) and metastasis-free survival (MFS; P = 0.015 and P < 0.001). Multivariate analysis revealed prognostic independence between HER-2/neu and the uPA/PAI-1 axis for DFS and MFS. Both uPA and PAI-1 had no significant discriminatory effect among HER-2/neu-positive patients for DFS. The prognostic value of HER-2/neu overexpression for MFS, however, was significantly enhanced by elevated uPA expression (P = 0.053). Stable transfection of the HER-2/neu gene into multiple human breast cancer cell lines resulted in consistent down-regulation of uPA or PAI-1 expression. In addition, anti-HER-2/neu antibodies did not significantly affect uPA or PAI-1 expression in human cancer cell lines naturally overexpressing HER-2/neu. CONCLUSIONS: The present findings suggest that the invasive phenotype elicited by HER-2/neu overexpression in breast cancer is not a direct effect of uPA or PAI-1 expression. HER-2/neu and the uPA/PAI-1 axis have been shown to affect the invasive capacity of breast cancer independently. Determination of uPA can provide significant additional prognostic information for MFS in HER-2/neu-positive and -negative patients.


Sujet(s)
Tumeurs du sein/anatomopathologie , Inhibiteur-1 d'activateur du plasminogène/analyse , Récepteur ErbB-2/analyse , Activateur du plasminogène de type urokinase/analyse , Adulte , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux humanisés , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Survie sans rechute , Test ELISA , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Vecteurs génétiques/génétique , Humains , Adulte d'âge moyen , Analyse multifactorielle , Métastase tumorale , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Récepteur ErbB-2/génétique , Retroviridae/génétique , Trastuzumab , Cellules cancéreuses en culture
12.
Clin Cancer Res ; 7(6): 1743-9, 2001 Jun.
Article de Anglais | MEDLINE | ID: mdl-11410515

RÉSUMÉ

PURPOSE: Urokinase-type plasminogen activator (uPA) and its inhibitor, plasminogen activator inhibitor (PAI)-1, have been shown to be related to poor prognosis in a variety of malignant solid tumors. Studies on the prognostic relevance of uPA and PAI-1 in ovarian cancer, however, have been inconclusive. The current study tests the hypothesis that elevated expression of uPA and PAI-1 is associated with prognosis and disease progression. EXPERIMENTAL DESIGN: uPA and PAI-1 were prospectively measured by quantitative ELISA in tumor samples from 103 ovarian cancer patients (82 primary invasive epithelial carcinomas, 9 low malignant potential tumors, and 12 recurrent ovarian carcinomas). RESULTS: uPA but not PAI-1 levels were consistently associated with malignant progression, with levels increased from low malignant potential tumors to primary tumors (uPA, P = 0.04; PAI-1, P = 0.019), from early to advanced disease stages (uPA, P = 0.014; PAI-1, P = 0.23), and from primary to intra-abdominal metastatic tumors (uPA, P = 0.001; PAI-1, P = 0.16). High uPA and PAI-1 levels were associated with residual tumor volumes of >1 cm (P = 0.001 and P = 0.016, respectively). Among invasive International Federation of Gynecologists and Obstetrician stages I-IV tumors, elevated levels of uPA (>5.5 ng/mg) and PAI-I (>18.8 ng/ml) were associated with a shortened progression-free survival (uPA, P = 0.003; PAI-1, P = 0.039) and overall survival (uPA, P = 0.0002; PAI-1, P = 0.007). In multivariate analysis, uPA retained prognostic independence for progression-free survival (P = 0.037) and overall survival (P = 0.006). CONCLUSIONS: These data suggest that the uPA/PAI-1 axis may play an important role in the intra-abdominal spread and reimplantation of ovarian cancer cells. The prognostic relevance of uPA and PAI-1 supports their possible role in the malignant progression of ovarian cancer.


Sujet(s)
Évolution de la maladie , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Inhibiteur-1 d'activateur du plasminogène/biosynthèse , Pronostic , Activateur du plasminogène de type urokinase/antagonistes et inhibiteurs , Activateur du plasminogène de type urokinase/biosynthèse , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Survie sans rechute , Activation enzymatique , Test ELISA , Femelle , Humains , Adulte d'âge moyen , Analyse multifactorielle , Tumeurs de l'ovaire/diagnostic , Récidive , Facteurs temps
13.
Oncologist ; 6 Suppl 3: 22-5, 2001.
Article de Anglais | MEDLINE | ID: mdl-11346681

RÉSUMÉ

Randomized controlled studies have demonstrated that both docetaxel and Herceptin are capable of increasing survival in patients with metastatic breast cancer. The two agents show synergy in vitro, and their use in combination is not likely to be associated with the problem of enhanced cardiotoxicity. In two trials of Herceptin plus docetaxel in patients with advanced breast cancer, preliminary data are available for 35 patients. These early results show that the combination is well-tolerated. No symptomatic cardiotoxicity has occurred. The preliminary response rates (RR) in these first- and second-line patients are 44% in one study and 63% in the other. In the subgroups of patients who were HER-2 3+ overexpressers, the RRs are currently 55% and 73%. In an attempt to maximize the efficacy of Herceptin, its use has also been studied in combination with docetaxel and a platinum salt, producing a preliminary RR of 78% in patients positive for HER-2 on the fluorescence in situ hybridization assay. These data are sufficiently promising to justify a study of the role of Herceptin in combination with adjuvant chemotherapy regimens containing docetaxel or docetaxel plus a platinum. The combination of Herceptin with adjuvant therapy containing docetaxel and a platinum may provide a helpful alternative to the potentially cardiotoxic Herceptin/anthracycline-containing regimens currently under investigation.


Sujet(s)
Antinéoplasiques/pharmacologie , Paclitaxel/analogues et dérivés , Taxoïdes , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Docetaxel , Association de médicaments , Prévision , Humains , Paclitaxel/administration et posologie , Trastuzumab
14.
Semin Oncol ; 28(1 Suppl 3): 13-9, 2001 Feb.
Article de Anglais | MEDLINE | ID: mdl-11301370

RÉSUMÉ

The discovery of the HER2/neu proto-oncogene and its role in the pathogenesis of breast cancer tumors, and the development of the anti-HER2 monoclonal antibody, trastuzumab (Herceptin; Genentech, South San Francisco, CA), directed against the HER2 receptor represent major milestones in the research developments in breast cancer, making trastuzumab the first monoclonal antibody available for treatment of this disease. Clinical trials in HER2-positive patients have demonstrated that the combined use of targeted therapy with trastuzumab in conjunction with cytotoxic chemotherapy is associated with improved time to disease progression and overall survival. Unfortunately, findings also demonstrate an increased risk for cardiotoxicity when trastuzumab is combined with anthracyclines. For HER2/neu-overexpressing breast cancer patients, the adjuvant use of trastuzumab will become paramount; therefore, it must be evaluated in a randomized controlled trial. There is disagreement regarding the design of such a trial, largely because of the ubiquitous use of anthracyclines in the adjuvant setting and the opposing necessity of avoiding anthracycline plus trastuzumab combinations. Combination index values for various chemotherapeutic drugs in combination with trastuzumab demonstrate dramatic synergistic interactions with the platinum agents and with docetaxel (Taxotere; Aventis Pharmaceuticals, Inc, Parsippany, NJ). The greatest level of synergy has been demonstrated with the triple-drug combination of docetaxel, platinum, and trastuzumab in which synergy is demonstrated, even at low doses. The adjuvant trial design for the Breast Cancer International Research Group uses a control arm of doxorubicin/cyclophosphamide for four cycles followed by docetaxel for four cycles and the second arm contains the addition of trastuzumab to the taxane sequence. The third arm, a non-anthracycline-containing regimen, contains docetaxel, a platinum agent (either cisplatin or carboplatin), and trastuzumab. The rationale for the selection of this three-drug regimen is based on the biology of the system and preclinical and clinical findings that demonstrate a high potential for clinical synergy.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Paclitaxel/analogues et dérivés , Taxoïdes , Anticorps monoclonaux/effets indésirables , Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Tumeurs du sein/génétique , Cisplatine/administration et posologie , Essais cliniques comme sujet , Docetaxel , Synergie des médicaments , Gènes erbB-2 , Défaillance cardiaque/induit chimiquement , Humains , Paclitaxel/administration et posologie , Pronostic , Proto-oncogène Mas , Trastuzumab
15.
N Engl J Med ; 344(11): 783-92, 2001 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-11248153

RÉSUMÉ

BACKGROUND: The HER2 gene, which encodes the growth factor receptor HER2, is amplified and HER2 is overexpressed in 25 to 30 percent of breast cancers, increasing the aggressiveness of the tumor. METHODS: We evaluated the efficacy and safety of trastuzumab, a recombinant monoclonal antibody against HER2, in women with metastatic breast cancer that overexpressed HER2. We randomly assigned 234 patients to receive standard chemotherapy alone and 235 patients to receive standard chemotherapy plus trastuzumab. Patients who had not previously received adjuvant (postoperative) therapy with an anthracycline were treated with doxorubicin (or epirubicin in the case of 36 women) and cyclophosphamide alone (138 women) or with trastuzumab (143 women). Patients who had previously received adjuvant anthracycline were treated with paclitaxel alone (96 women) or paclitaxel with trastuzumab (92 women). RESULTS: The addition of trastuzumab to chemotherapy was associated with a longer time to disease progression (median, 7.4 vs. 4.6 months; P<0.001), a higher rate of objective response (50 percent vs. 32 percent, P<0.001), a longer duration of response (median, 9.1 vs. 6.1 months; P<0.001), a lower rate of death at 1 year (22 percent vs. 33 percent, P=0.008), longer survival (median survival, 25.1 vs. 20.3 months; P=0.01), and a 20 percent reduction in the risk of death. The most important adverse event was cardiac dysfunction of New York Heart Association class III or IV, which occurred in 27 percent of the group given an anthracycline, cyclophosphamide, and trastuzumab; 8 percent of the group given an anthracycline and cyclophosphamide alone; 13 percent of the group given paclitaxel and trastuzumab; and 1 percent of the group given paclitaxel alone. Although the cardiotoxicity was potentially severe and, in some cases, life-threatening, the symptoms generally improved with standard medical management. CONCLUSIONS: Trastuzumab increases the clinical benefit of first-line chemotherapy in metastatic breast cancer that overexpresses HER2.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Récepteur ErbB-2/immunologie , Adulte , Sujet âgé , Anthracyclines/effets indésirables , Anthracyclines/usage thérapeutique , Anticorps monoclonaux/effets indésirables , Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Tumeurs du sein/métabolisme , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Cyclophosphamide/effets indésirables , Cyclophosphamide/usage thérapeutique , Évolution de la maladie , Doxorubicine/effets indésirables , Doxorubicine/usage thérapeutique , Épirubicine/usage thérapeutique , Femelle , Cardiopathies/induit chimiquement , Humains , Adulte d'âge moyen , Métastase tumorale , Paclitaxel/effets indésirables , Paclitaxel/usage thérapeutique , Récepteur ErbB-2/métabolisme , Analyse de survie , Trastuzumab
16.
Biochem Pharmacol ; 61(2): 179-89, 2001 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-11163332

RÉSUMÉ

The in vivo administration of enzyme-inhibiting drugs for cancer and infectious disease often results in overexpression of the targeted enzyme. We have developed an enzyme-catalyzed therapeutic agent (ECTA) approach in which an enzyme overexpressed within the resistant cells is recruited as an intracellular catalyst for converting a relatively non-toxic substrate to a toxic product. We have investigated the potential of the ECTA approach to circumvent the thymidylate synthase (TS) overexpression-based resistance of tumor cells to conventional fluoropyrimidine [i.e. 5-fluorouracil (5-FU)] cancer chemotherapy. (E)-5-(2-Bromovinyl)-2'-deoxy-5'-uridyl phenyl L-methoxyalaninylphosphoramidate (NB1011) is a pronucleotide analogue of (E)-5-(2-bromovinyl)-2'-deoxyuridine (BVdU), an antiviral agent known to be a substrate for TS when in the 5'-monophosphorylated form. NB1011 was synthesized and found to be at least 10-fold more cytotoxic to 5-FU-resistant, TS-overexpressing colorectal tumor cells than to normal cells. This finding demonstrates that the ECTA approach to the design of novel chemotherapeutics results in compounds that are selectively cytotoxic to tumor cell lines that overexpress the target enzyme, TS, and therefore may be useful in the treatment of fluoropyrimidine-resistant cancer.


Sujet(s)
Antinéoplasiques/métabolisme , Broxuridine/analogues et dérivés , Broxuridine/métabolisme , Conception de médicament , Promédicaments/métabolisme , Thymidylate synthase/métabolisme , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacologie , Broxuridine/administration et posologie , Broxuridine/pharmacologie , Nucléotides désoxyuridyliques/composition chimique , Nucléotides désoxyuridyliques/métabolisme , Modèles animaux de maladie humaine , Tests de criblage d'agents antitumoraux , Humains , Souris , Souris nude , Promédicaments/administration et posologie , Promédicaments/pharmacologie , Thymidylate synthase/administration et posologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Breast Cancer Res Treat ; 69(1): 53-63, 2001 Sep.
Article de Anglais | MEDLINE | ID: mdl-11759828

RÉSUMÉ

Available clinical and experimental data on the effect of HER-2/neu overexpression on chemosensitivity are controversial. It was the purpose of this in vitro study to define the association between HER-2/neu overexpression and the sensitivity to the chemotherapeutic drug combinations of cyclophosphamide, methotrexate and 5-fluorouracil (CMF) and 5-fluorouracil, epirubicin and cyclophosphamide (FEC) of breast cancer cells derived from 140 chemotherapy-naïve patients at the time of primary surgery. Both drug combinations were tested at six different concentrations ranging from 6.25-200% peak plasma concentration (PPC). Immunohistochemical detection of HER-2/neu overexpression was performed with the HER-2/neu antibodies, CB11, TAB250 and AO485, in the same tumor specimens. Immunoreactions were determined as negative (0/1+), weakly positive (2+) and strongly positive (3+). However, the antibodies varied in their degrees of sensitivity. Breast cancer samples with strong (3+) HER-2/neu overexpression demonstrated 90% growth inhibition (IC90) at significantly lower PPC values, using the CB11 (p = 0.048), TAB250 (p = 0.007) and AO485 (p < or =0.01) antibodies, and showed 50% growth inhibition (IC50) at significantly lower PPC values, using the CB11 antibody (p = 0.01) compared to their counterparts with lower levels of HER-2/neu expression. When analyzing the group of patients with intermediate and strong HER-2/neu overexpression (2+ and 3+), an association between HER-2/neu overexpression and increased chemosensitivity was seen with the TAB250 (p = 0.044) and AO485 (p = 0.032) antibodies, but not with the CB11 antibody (p =0.8) at the IC90 level. Differences in chemosensitivity between samples with strong HER-2/neu overexpression and those with lower levels were then analyzed separately for CMF and FEC. Both regimens achieved 90% tumor growth inhibition at lower PPC values in samples with strong HER-2/neu overexpression (3+) compared to their counterparts with lower expression levels (AO485 p = 0.011 for CMF, and p = 0.09 for FEC). Cumulative concentration-response plots of tumors responding in vitro with 90% tumor cell inhibition showed a stronger dose dependence for both CMF and FEC among tumor samples with strong HER-2/neu overexpression compared to those with lower levels of expression. In conclusion, the data show that HER-2/neu overexpression was not associated with in vitro drug resistance to CMF or FEC. In contrast, tumors with strong HER-2/neu overexpression demonstrated increased dose-dependent in vitro sensitivity to both the FEC and CMF regimens.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Régulation de l'expression des gènes tumoraux , Récepteur ErbB-2/biosynthèse , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Tumeurs du sein/anatomopathologie , Cyclophosphamide/administration et posologie , Relation dose-effet des médicaments , Résistance aux médicaments antinéoplasiques , Tests de criblage d'agents antitumoraux , Épirubicine/administration et posologie , Femelle , Fluorouracil/administration et posologie , Humains , Immunohistochimie , Méthotrexate/administration et posologie , Adulte d'âge moyen , Régulation positive
18.
Clin Breast Cancer ; 2 Suppl 1: S15-9, 2001 Oct.
Article de Anglais | MEDLINE | ID: mdl-11970740

RÉSUMÉ

Overexpression of the p185/HER2 protein is seen in 20%-25% of primary breast cancers and is associated with poor prognosis. Recent phase II and III clinical trials demonstrate that trastuzumab is active against breast tumors, both as a single agent and in combination with chemotherapy. In patients with HER2-overexpressing metastatic breast cancer, use of trastuzumab in combination with chemotherapy is associated with a 20% reduction in relative risk of death and an increase in median survival from 20.3 to 25.1 months compared to chemotherapy alone. Side effects include fever and chills and an unexpected increase in doxorubicin/trastuzumab-associated cardiomyopathy. Clinical development is now focused on trastuzumab in combination with chemotherapy regimens that do not contain an anthracycline. Trastuzumab in combination with docetaxel is synergistic in vitro. Data from ongoing clinical trials are consistent with this finding. Preliminary data from 3 phase II studies suggest a 44%-63% response rate when the combination is used first or second line in HER2-overexpressing metastatic breast cancer. The combination of docetaxel with trastuzumab is well tolerated and has not been associated with significant cardiotoxicity. Given in vitro evidence that platinum salts act synergistically with trastuzumab and docetaxel, and phase II data suggesting clinical efficacy and good tolerability, the combination of platinum salt plus trastuzumab and docetaxel is now being assessed in adjuvant trials


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Taxoïdes/usage thérapeutique , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Tumeurs du sein/chirurgie , Traitement médicamenteux adjuvant , Essais cliniques comme sujet , Docetaxel , Femelle , Humains , Hybridation fluorescente in situ , Récepteur ErbB-2/biosynthèse , Analyse de survie , Taxoïdes/administration et posologie , Taxoïdes/effets indésirables , Trastuzumab
19.
Semin Oncol ; 27(5 Suppl 9): 13-9, 2000 Oct.
Article de Anglais | MEDLINE | ID: mdl-11049052

RÉSUMÉ

The physical characteristics of tumor antigens that would make the most ideal targets for antibody therapeutics include cell surface expression; high, stable expression levels in tumor cells; low or absent expression in normal tissues; lack of a soluble form of the antigenic target; and lack of internalization of the antigen/antibody complex. HER2/neu is a 185-kd surface membrane protein that is overexpressed in approximately 25% of human breast cancers due to amplification of the HER2 gene. Overexpression of the gene results in ligand-independent activation of HER2 kinase, causing mitogenic signal transduction and increased cell proliferation. Consequently, patients with this alteration have a worse clinical prognosis. Trastuzumab (Herceptin; Genentech, Inc, So. San Francisco, CA), a humanized anti-HER2 monoclonal antibody, has significant clinical activity against metastatic breast cancers with HER2/neu overexpression, despite the fact that the p185HER2 protein product lacks some of the ideal characteristics of tumor antigens listed above. We propose that the efficacy of trastuzumab may be explained on the basis of its effects on signal transduction, which is independent from its immune mechanism(s) of action. Furthermore, trastuzumab is synergistic with some chemotherapeutic drugs, resulting in improved therapeutic efficacy. Thus, in the case of trastuzumab, a clear distinction may be drawn between the use of monoclonal antibodies as immuneactive agents and their use to achieve a desired cellular/biochemical activity.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Récepteur ErbB-2 , Anticorps monoclonaux humanisés , Tumeurs du sein/métabolisme , Tumeurs du sein/physiopathologie , Humains , Trastuzumab
20.
Cancer Treat Res ; 103: 57-75, 2000.
Article de Anglais | MEDLINE | ID: mdl-10948442
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...