Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Urol Oncol ; 31(8): 1567-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22608542

ABSTRACT

OBJECTIVES: Overexpression of epidermal growth factor receptor (EGFR) and angiogenic factors is associated with the progression of androgen-independent prostate cancer (AIPC). We examined the effects of vandetanib, an inhibitor of vascular endothelial growth factor (VEGFR), EGFR, and rearranged during transfection (RET) tyrosine-kinase activities, alone or combined with docetaxel, on PC3 docetaxel-sensitive (PC3wt) or docetaxel-resistant (PC3R) AIPC cell growth in vivo and in vitro. METHODS: Mice bearing PC3wt or PC3R tumors were treated for 3 weeks with vandetanib (25 or 50 mg/kg/d p.o., 5 d/wk), docetaxel (10 or 30 mg/kg i.p., 1 d/wk), or their combination (low or high doses). Xenograft tumors were analyzed for expression of Ki-67, EGFR, VEGFR2, and production of VEGFA. RESULTS: On PC3wt, vandetanib at both doses stimulated tumor growth, whereas docetaxel at both doses exerted strong growth-inhibiting effects. The low-dose vandetanib-docetaxel combination resulted in tumor growth similar to that of control, whereas the high-dose combination induced a significant antiproliferative effect. In contrast, on PC3R, the low-dose of vandetanib had no effect on tumor growth, whereas the high-dose of vandetanib significantly inhibited tumor growth. Docetaxel at both doses exerted moderate and transient antitumor effects. The combination of high-dose vandetanib with high-dose docetaxel resulted in antiproliferative effects, which were lower than expected from the sum of individual drug effects. Importantly, tumor analyses revealed overexpression of the EGFR/VEGFR pathways in PC3R relative to PC3wt. CONCLUSION: Present results suggest that vandetanib should not be associated with docetaxel in treatment-naive or docetaxel-resistant prostate cancer (CaP). The use of high-dose vandetanib alone may warrant further investigation in patients with docetaxel-resistant AIPC overexpressing VEGFR/EGFR pathways.


Subject(s)
Piperidines/pharmacology , Prostatic Neoplasms/drug therapy , Quinazolines/pharmacology , Taxoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred Strains , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
2.
Invest New Drugs ; 31(1): 59-65, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22714791

ABSTRACT

BACKGROUND: Radiotherapy (RT) is the standard treatment for uveal melanoma. However it can cause damage to the retina and optic nerve. This study examined the in vitro and in vivo effects of the anti-VEGF monoclonal antibody bevacizumab associated with radiotherapy (RT) on tumor growth and tumor proliferation and vasculature on OCM-1 human uveal melanoma cell line. METHODS: The anti-proliferative effects of bevacizumab, RT and their combination were tested both in vitro (OCM-1 cells co-cultured with HUVEC cells in Transwell plates) and in vivo (OCM-1 tumor xenografts in nude mice). In addition, treatment effects in vitro on VEGF secretion, as well as treatment effects in vivo on tumor proliferation (Ki67 labelling), tumor vasculature (VEGFR2 labelling) and VEGF tumoral concentration were analyzed. RESULTS: Bevacizumab given alone had a significant impact on tumor growth in vivo (and moderate effects in vitro). The bevacizumab-RT combination had additive effects in vitro (tumor cell proliferation) and in vivo (tumor growth), which translated into a significant decrease in Ki67 expression, VEGFR2 labelling and VEGF tumoral content. CONCLUSIONS: The bevacizumab-RT combination could be a promising clinical option to explore for the management of human uveal melanoma, since it may allow RT dose reduction without loss of antitumor efficacy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Melanoma/drug therapy , Melanoma/radiotherapy , Uveal Neoplasms/drug therapy , Uveal Neoplasms/radiotherapy , Animals , Bevacizumab , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Female , Human Umbilical Vein Endothelial Cells , Humans , Melanoma/pathology , Mice , Mice, Nude , Tumor Burden/drug effects , Uveal Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
3.
Urol Oncol ; 31(6): 776-86, 2013 Aug.
Article in English | MEDLINE | ID: mdl-21795074

ABSTRACT

OBJECTIVE: To examine the anti-proliferative effect of the combination of docetaxel, the cornerstone of modern chemotherapy for prostate cancer, and vandetanib, a potent inhibitor of VEGFR-2 tyrosine kinase, applied to the representative hormone-refractory human prostate cancer cell line PC3. The aim is to analyze if a supra-additive/synergic effect of the combined treatment on cell viability exists and to understand the molecular key-factors involved. We first hypothesized an effect of vandetanib in modulation the function of MDR1, leading to a longer retention of docetaxel inside the cell. It may also be possible that vandetanib could modulate the docetaxel-induced changes in expression of prosurvival and proapoptotic proteins, leading to a positive balance forward cell death. MATERIALS AND METHODS: We used PC3 cells either wild type (PC3wt) or with acquired resistance to docetaxel (PC3R), characterized by a higher expression of MDR1. We studied both mRNA and protein, the expression of EGF and VEGF receptors at a basal level and after each treatment, as well as the expression of cell cycle and apoptosis related genes. RESULTS: Cell proliferation data suggested a supra-additive cytotoxic effect of the combination of docetaxel plus vandetanib, when given together or with the sequence vandetanib followed by docetaxel. We did not observe any effect of vandetanib on MDR1, in the PC3R cell lines, characterized by a higher pump expression than PC3wt. On the other side, we defined a number of key factors involved in the pro- and anti-survival balance, which regulation, by single drugs and/or by combined treatment, could explain the effect on cell cytotoxicity; also where there are apparently contradictory results. CONCLUSIONS: Our data suggest that combined treatment with vandetanib and docetaxel alters the balance of proapoptotic and prosurvival proteins, ultimately leading to potentiation of docetaxel-induced apoptosis in human prostate cancer cells in vitro, irrespective of cells being sensitive or resistant to docetaxel.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Piperidines/administration & dosage , Prostatic Neoplasms, Castration-Resistant/metabolism , Quinazolines/administration & dosage , Taxoids/administration & dosage , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cell Survival , Coloring Agents/pharmacology , Docetaxel , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Humans , Male , Tetrazolium Salts , Thiazoles , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
Fundam Clin Pharmacol ; 26(4): 530-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21623901

ABSTRACT

The combination of lapatinib and capecitabine is approved in Her2+ metastatic breast cancer. However, the pharmacological mechanisms for this association have not been fully elucidated. In this non-clinical study, we evaluated the efficacy of this association on a panel of six human breast cancer cell lines as a means to identify the molecular determinants of response to this combination. Cell viability was evaluated after concomitant/sequential exposure, and response/resistance determinants for each drug such as dihydropyrimidine dehydrogenase (DPD), thymidylate synthase (TS), thymidine phosphorylase, Bax, Bcl2, P21 levels, and phospho p42/44 and HER1/2 signaling pathway were studied. Lapatinib proved to markedly downregulate TS activity, thus suggesting a subsequent better efficacy of capecitabine. Capecitabine optimized the downregulation of p-AKT and p-P42/44 expression by lapatinib. Consequently, we observed an increase in the Bax/Bcl2 ratio and p21 protein expression in cells exposed to the combination. Overall, our data showed that whatever the schedule and the cell line were, additive to synergistic interaction was achieved in our models. The optimal in vitro combination was finally tested in tumor-bearing mice. Our results fully confirmed that associating both drugs led to a 77% reduction in tumor growth as compared with control animals in BT474-xenografted models. Taken together, this non-clinical study shows that lapatinib and capecitabine modulate each other's molecular determinants of response and that concomitant dosing seems to be the optimal way to combine these drugs. Besides, modulation of TS expression by lapatinib makes its association with capecitabine a promising way to overcome breast cancers resistant in relation with TS overexpression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Quinazolines/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Capecitabine , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Dihydrouracil Dehydrogenase (NADP)/metabolism , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Humans , Lapatinib , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/administration & dosage , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Thymidine Phosphorylase/metabolism , Thymidylate Synthase/metabolism , Xenograft Model Antitumor Assays
5.
Oral Oncol ; 47(10): 940-5, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21862388

ABSTRACT

BACKGROUND: Cetuximab (Cet), an anti-epidermal growth factor receptor (EGFR) monoclonal antibody, is a standard of care in advanced head and neck (HN) cancer when combined with irradiation or cisplatin. The TPF association combining docetaxel (T), cisplatin (P) and fluorouracil (F) has become a chemotherapy of reference in the management of HN tumors. The aim of the study was to evaluate the anti-tumor activity of the association of Cet with TPF. In addition, the potential benefit from adding bevacizumab (Bev), an anti-VEGF monoclonal antibody, was examined. METHODS: Investigations were performed on CAL33 human head and neck cancer cell line (high content of EGFR) injected as an orthotopic xenograft into the mouth floor of nude mice. The anti-tumor efficacy of Cet (2.5 mg/kg), Bev (20 mg/kg) and of TPF (at the MTD: T 20 mg/kg, P 6 mg/kg, F 17 mg/kg), administered alone or in combination on day 3 and day 10 after tumor cell injection, was assessed on day 12 (animal sacrifice for ethical reasons, 10 animals per treatment group). Tumor volume, tumor histology, tumor cell proliferation (Ki67, immunochemistry) and apoptosis (Bcl2, immunochemistry) were examined. RESULTS: As compared to controls, TPF-Cet and TPF-Cet-Bev had a significant impact on tumor volume and histological response. The highest efficacy was observed with the TPF-Cet combination. The effects of the TPF-Cet-Bev association were not significantly different from those observed with TPF-Cet. Data were corroborated by a diminution in Ki67 labeling and Bcl2 expression. CONCLUSION: The association of Cet with TPF can be considered a beneficial clinical option in advanced HN cancer patients.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Head and Neck Neoplasms/drug therapy , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Apoptosis/drug effects , Bevacizumab , Cell Line, Tumor , Cetuximab , Cisplatin/administration & dosage , Docetaxel , Drug Therapy, Combination/methods , Fluorouracil/administration & dosage , Head and Neck Neoplasms/pathology , Humans , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Taxoids/administration & dosage , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Oral Oncol ; 47(5): 340-4, 2011 May.
Article in English | MEDLINE | ID: mdl-21421338

ABSTRACT

We previously reported on head and neck tumor xenografts that the tumor regression induced by a triple combination of irradiation (RT), anti-EGFR and anti-angiogenic therapies was followed, after treatment arrest, by tumor re-growth characterized by activation of the AKT signaling pathway. Since mTOR is the main AKT-related messenger, the aim of this study was to add the mTOR inhibitor temsirolimus to a tri-therapy with RT plus anti-EGFR and anti-angiogenic drugs in order to improve anti-tumor effects. The human head and neck cancer cell line CAL33 (over-expressing EGFR and secreting VEGF-A) was xenografted in nude mice. Treatment (20 mice per treatment group) was administered for 2 weeks and consisted of either vehicle (control), temsirolimus (5mg/kg i.p. five times a week), tri-therapy with RT (6 Gy three times a week) combined with cetuximab (0.5mg/kg i.p. five times a week) and bevacizumab (5mg/kg i.p. five times a week) or the temsirolimus-tri-therapy association. The time to reach a tumor volume of 2000 mm(3) was significantly different between the four treatment groups (Log Rank p<0.0001), with a median of 29.5, 44.5, 67.0 and 70.0 days for control, temsirolimus, tri-therapy and combination groups, respectively. The combination of temsirolimus plus tri-therapy produced the longest growth-inhibiting effects (tri-therapy versus combination, p=0.01). No significant interaction was observed between temsirolimus and the tri-therapy, suggesting that temsirolimus, on the one hand, and RT-cetuximab-bevacizumab, on the other, exert additive effects on tumor growth inhibition. These decreases observed on tumor growth were corroborated by the parallel decreases observed on tumor proliferation (Ki67) and on anti-apoptotic markers (Bcl2). These results suggest that temsirolimus exhibits synergistic antiproliferative effects when administered in combination with irradiation, anti-EGFR and anti-angiogenic therapies in head and neck cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/drug effects , Animals , Antibodies, Monoclonal, Humanized , Bevacizumab , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cetuximab , Combined Modality Therapy/methods , Drug Synergism , Head and Neck Neoplasms/metabolism , Humans , Mice , Mice, Nude , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
7.
Oral Oncol ; 46(3): 172-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20156700

ABSTRACT

Head-and-neck (H&N) tumor re-growth is clinically observed after arrest of anti-EGFR therapies. In the present study, we compared, for a similar dose exposure to anti-EGFR therapies, constant (CS) with tapered (TS) schedules (i.e. progressive dose reduction) for cetuximab, a mAb, and gefitinib, a TKI. Mice bearing CAL33 H&N tumors with high EGFR content were treated with cetuximab or gefitinib. CS consisted of 7 days of cetuximab or gefitinib administration and TS of 7 days of drugs followed by 7 days of administration at decreased doses (divided by 2 every day). Gefitinib TS but not CS slowed down tumor re-growth. Cetuximab TS had a stronger and longer-lasting effect than CS, paralleled by a down-regulation of EGFR expression. Whatever the drug, TS decreased tumor re-growth more efficiently than CS. This new drug schedule could be of interest in the management of anti-EGFR based therapies in H&N cancer.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Quinazolines/administration & dosage , Animals , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Blotting, Western , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab , Female , Gefitinib , Head and Neck Neoplasms/pathology , Mice , Mice, Nude , Transplantation, Heterologous
8.
Pharmaceuticals (Basel) ; 3(7): 2238-2247, 2010 Jul 19.
Article in English | MEDLINE | ID: mdl-27713352

ABSTRACT

The incidence of prostate cancer increases with age and because of its high prevalence this disease has become a major public health concern. Despite advances in our understanding of the biological mechanisms responsible for the development of this cancer, the transition to the hormone refractory stage (HRPC) and metastatic progression pose real problems of clinical management. Currently, docetaxel chemotherapy has been shown to have a slight but significant impact on survival, though the gain in median survival is still less than three months. Research is therefore continuing to improve treatment outcomes. The progression of prostate cancer is accompanied by the overexpression of EGFR (epidermal growth factor receptor) in a very large majority of cases, suggesting that this may play a mechanistic role. Unfortunately, although preclinical findings seem to be promising for therapies targeting the EGFR in HRPC, current clinical results are disappointing. These results should however encourage us to look for different ways of using anti-EGFR agents or combining them with other targeted therapies.

9.
Expert Opin Emerg Drugs ; 14(2): 299-310, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19519286

ABSTRACT

Current development of molecular targeted therapies in oncology is particularly active. The aim of this study is to review recent advances in the field of molecular targeted therapies for head and neck squamous cell carcinoma (HNSCC). As EGFR signaling pathway and angiogenesis play a key role in the growth of HNSCC, EGFR with its downstream effectors and molecular factors implicated in the angiogenesis process, such as VEGF and its receptors, represent the main targets of the new therapeutic agents now in development. Today, cetuximab, an anti-EGFR monoclonal antibody, is the only targeted therapy approved for the treatment of HNSCC in patients with locally advanced tumors, in association with radiotherapy, and in patients with recurrent or metastatic diseases. Future progress is expected with the integration of cetuximab into induction chemotherapeutic regimens or in association with concurrent chemoradiotherapy for locally advanced tumors and with the development and evaluation of other molecular targeted therapies such as antiangiogenic drugs. As these innovative molecules start to be used in clinical practice, the identification of predictive markers for efficacy and toxicity becomes a crucial issue.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Drug Delivery Systems , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/therapeutic use , Cetuximab , Drug Delivery Systems/adverse effects , Drug Delivery Systems/standards , Drugs, Investigational/therapeutic use , Humans
10.
J Cancer Res Clin Oncol ; 135(11): 1615-23, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19499245

ABSTRACT

PURPOSE: Head and neck squamous cell carcinoma (HNSCC) represents an ideal model for assessing the impact of anemia and tumor hypoxia on the response to radiotherapy (RT). Various treatment strategies aimed at increasing tumor oxygenation in HNSCC patients have been studied and these studies have been fueled by evidence that hypoxia and, unexpectedly, erythropoietin (EPO), adversely affect the radiosensitivity of cells. The purpose of the present study was to experimentally examine the relationship between hypoxia, EPO, its receptor EPOR, EGFR and their effects on the survival and radiosensitivity of HNSCC cells underwent hypoxia. METHODS: We used Cal-166 head and neck cell line to investigate different cellular responses after RT given in oxic and hypoxic conditions, focusing on the role of EPO administration in cell proliferation and in regulating response to RT. RESULTS: Our results show that EPO do not evoke a physiologic response on EPOR-bearing tumor cells as assessed by cellular growth and proliferation. In addition, we present some indications that EPO could activate opposite signals related to proliferation, DNA repair and apoptosis. Among them, EGFR and AKT phosphorylation may play a role in radioresistance. CONCLUSIONS: We concluded that the expression of the EPOR in Cal-166 cells does not seem essential for their growth and that administration of EPO does not affect RT efficacy.


Subject(s)
Cell Hypoxia , Erythropoietin/pharmacology , Head and Neck Neoplasms/radiotherapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/radiation effects , Cytoprotection , DNA Repair , DNA-Binding Proteins/physiology , ErbB Receptors/analysis , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Proto-Oncogene Proteins c-akt/physiology , Receptors, Erythropoietin/analysis , X-ray Repair Cross Complementing Protein 1
11.
Eur J Cancer ; 44(13): 1922-30, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18691881

ABSTRACT

The aim of this study was to examine the anti-tumour effects of dual vertical VEGF targeting consisting in the association between bevacizumab, a VEGF-depleting drug, and the VEGF receptor antityrosine kinase AZD2171. Mice bearing human head and neck CAL33 xenografted tumours were treated once daily for 11 d with either vehicle (controls), AZD2171 (2.5mg/kg/day, p.o.), bevacizumab (5mg/kg/day, i.p.) or the bevacizumab-AZD2171 combination. The AZD2171-bevacizumab combination produced additive effects on tumour growth and reduced the number of proliferating cells relative to control. Bevacizumab did not influence tumour vascular necrosis whilst AZD2171 (p=0.01) and the combination (p=0.01) increased it. The number of mature tumour cells decreased significantly with the combination treatment only (p=0.001), which induced the largest increase in the Bax/Bcl2 ratio (up to 25-fold) and a progressive 3-fold decrease in HIF1-alpha expression between 24h and 192h. The present data indicate that there is no redundancy in targeting the same angiogenic pathway with the presently tested clinically applicable drugs. The study provides a strong rationale for future clinical trials.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Neovascularization, Pathologic/prevention & control , Quinazolines/therapeutic use , Tongue Neoplasms/drug therapy , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Bevacizumab , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Combinations , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Tongue Neoplasms/blood supply , Tongue Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
12.
Acta Oncol ; 47(8): 1502-12, 2008.
Article in English | MEDLINE | ID: mdl-18607838

ABSTRACT

BACKGROUND: The gene encoding cortactin, CTTN (locus 11q13), an actin-binding substrate of Src kinases, is frequently amplified in breast and head and neck squamous cell carcinomas (HNSCC) and cortactin overexpression is thought to contribute in a significant way to the invasive phenotype of these tumors. Elevated Epidermal Growth Factor receptor (EGFR) expression is also commonly observed in HNSCC and has been associated with poor prognosis and resistance to cytotoxic agents, including ionizing radiation. It has been suggested that cortactin overexpression may increase EGFR levels in these tumors by affecting receptor downregulation, however we recently found by multivariate analysis, that cortactin expression status remained an independent prognostic factor for local recurrence, disease-free survival, and overall survival. MATERIAL AND METHODS: To examine the potential link between cortactin overexpression and EGFR status, we compared cortactin and EGFR levels in a series of tumor lines derived from HNSCC. RNAi-mediated silencing was performed in cortactin overexpressing cells and in vivo tumoral potential with respect to cortactin and EGFR status was analyzed. RESULTS AND DISCUSSION: Cortactin and EGFR levels were not strictly coupled in these lines and cortactin depletion did not decrease steady state receptor levels, although it did affect the epithelial to mesenchymal phenotypic conversion of cells. These results, together with clinical findings point to the existence of an EGFR-independent role of cortactin in HNSCC that may have important implications regarding the design of targeted therapies to combat tumor spread.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cortactin/metabolism , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Animals , Blotting, Southern , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Cortactin/genetics , ErbB Receptors/genetics , Female , Fluorescent Antibody Technique , Gene Amplification , Head and Neck Neoplasms/genetics , Humans , Immunoenzyme Techniques , Kidney/metabolism , Kidney/pathology , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology
13.
BJU Int ; 102(5): 622-7, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18494832

ABSTRACT

OBJECTIVE: To evaluate the effects of the association between docetaxel and the somatostatin analogue lanreotide on the androgen-independent prostate cancer cell line PC3, either sensitive or made resistant to docetaxel (PC3R), as new drugs and new combinations have promising clinical activity in hormone-refractory prostate cancer. MATERIALS AND METHODS: We examined the effect of docetaxel and lanreotide on cell proliferation, with analysis of the mitogen-activated protein kinase pathway and expression of cell-cycle regulatory proteins. RESULTS: Combined treatment with docetaxel and lanreotide inhibited PC3 cell growth in vitro through an enhanced induction of cell death, compared with treatment with either agent alone; this result was particularly evident on PC3R cells. The results suggested that lanreotide could act as a P glycoprotein inhibitor in PC3R cells. CONCLUSION: The present results provide a promising therapeutic approach for using somatostatin analogues in hormone-refractory prostate cancer, in which lanreotide could interact with docetaxel in PC3R cells, with possible explanatory mechanisms which involve P glycoprotein-mediated docetaxel resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Blotting, Western , Cell Line, Tumor , Docetaxel , Drug Evaluation , Drug Resistance, Neoplasm , Humans , Male , Mitogen-Activated Protein Kinase Kinases/drug effects , Peptides, Cyclic/administration & dosage , Signal Transduction/drug effects , Somatostatin/administration & dosage , Somatostatin/analogs & derivatives , Taxoids/administration & dosage
14.
Crit Rev Oncol Hematol ; 62(1): 53-61, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17324578

ABSTRACT

Current development of targeted therapy in oncology is particularly active and concerns principally two types of agents which are monoclonal antibodies (Mabs) and tyrosine kinase inhibitors (TKIs). Epidermal growth factor receptor (EGFR) signaling pathways play a key role in the regulation of cell proliferation, survival and differentiation. Consequently, EGFR is one of the most-studied ligand-receptor systems and specific EGFR inhibition approaches are currently among the most promising and the most advanced in the clinical setting. Cetuximab (Erbitux), belonging to the Mabs family, gefitinib (Iressa) and erlotinib (Tarceva), belonging to the TKIs family, are among the most advanced anti-EGFR drugs at the clinical level. The aim of this review article is to compare at both experimental and clinical levels the key points which govern the activity of these two types of targeting agents.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Clinical Trials as Topic , Drug Delivery Systems/methods , Humans , Protein Kinase Inhibitors/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/drug effects
15.
Anticancer Drugs ; 17(7): 807-13, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16926630

ABSTRACT

The current reference treatment of hormone-refractory prostate cancer consists mainly of chemotherapy with docetaxel. To improve the management of advanced prostate cancer, one should examine the benefits of adding other agents to docetaxel. We examined the growth inhibitory effects of a triple combination, including the anti-epidermal growth factor receptor drug erlotinib, docetaxel and 5-fluoro-5'-deoxyuridine (the main intermediary metabolite of capecitabine), on the human prostate cancer cell lines PC3 and DU145, which are both devoid of androgen receptors. Marked synergistic cytotoxic effects were observed with the application of the double combination of erlotinib-5-fluoro-5'-deoxyuridine for both cell lines and to a lesser magnitude with the triple combination. For PC3 cells, all conditions resulted in synergistic interactions. The combination between erlotinib and docetaxel resulted in an approximately 50% reduction in thymidylate synthase activity (the molecular target of 5-fluorodeoxyuridine monophosphate, the active capecitabine anabolite) with an higher impact observed with DU145 cells than with PC3 cells. Neither erlotinib nor docetaxel alone displayed marked effects on thymidine phosphorylase activity (the enzyme that governs at the cellular level the final and crucial step in the activation cascade of capecitabine), in contrast to their combination that resulted in a strong increase in thymidine phosphorylase activity in PC3 cells. These data may serve as a rational basis for setting up clinical trials in advanced prostate cancer combining epidermal growth factor receptor-targeting agents like erlotinib together with docetaxel and capecitabine.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Floxuridine/therapeutic use , Prostatic Neoplasms/drug therapy , Quinazolines/therapeutic use , Taxoids/therapeutic use , Cell Line, Tumor , Cell Size , Cell Survival/drug effects , Docetaxel , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Erlotinib Hydrochloride , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Thymidine Phosphorylase/metabolism , Thymidylate Synthase/metabolism
16.
Curr Opin Oncol ; 18(4): 330-4, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16721126

ABSTRACT

PURPOSE OF REVIEW: This review aims to critically examine the preclinical background regarding the combination of drugs targeting the epidermal growth factor receptor and anti-angiogenic compounds. RECENT FINDINGS: There are studies exploring the anti-tumor efficacy of dual inhibitors, such as the compound ZD6474, which combines in the same molecule an anti-tyrosine kinase activity against the epidermal growth factor receptor and the vascular endothelial growth factor receptor. In addition, several studies have investigated the anti-tumor effects of combinations of an anti-epidermal growth factor receptor agent and a vascular endothelial growth factor receptor inhibitor. In general, in these studies, supra-additive anti-tumor efficacy was apparent when combining anti-epidermal growth factor receptor and anti-angiogenic treatments. Beneficial effects were also observed when combining this dual targeted therapy with either conventional chemotherapy or irradiation. SUMMARY: Early clinical trials combining the anti-epidermal growth factor receptor drug erlotinib (Tarceva) and the anti-angiogenic agent bevacizumab (Avastin) show acceptable toxicity and promising anti-tumor activity (lung cancer), which need to be confirmed in randomized trials.


Subject(s)
Angiogenesis Inhibitors/pharmacology , ErbB Receptors/metabolism , Neovascularization, Pathologic , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Humans , Piperidines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology
17.
Mol Cancer Ther ; 4(12): 1962-71, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16373711

ABSTRACT

Recent studies suggest the possibility of a direct antiangiogenic effect of anti-epidermal growth factor receptor (EGFR) drugs due to the presence of EGFR on endothelial cells. The aim of this study was to analyze the direct effect on endothelial cells of associating EGFR targeting, vascular endothelial growth factor receptor (VEGFR)-2 targeting, and irradiation. We examined both the cytotoxic effects and the effect on molecular markers resulting from the combined action of gefitinib (Iressa; anti-EGFR), ZM317450 [VEGFR tyrosine kinase inhibitor (VTKI); anti-VEGFR-2], and irradiation (radiation therapy) on HMME7 cells, an immortalized microvascular endothelial cell of human origin. The presence of a functional EGFR pathway sensitive to gefitinib was shown in HMME7 cells (gefitinib-induced decrease in phospho-EGFR, phospho-p42/p44, and phospho-Akt). The stimulation of VEGFR-2 pathway led to an increase in Akt phosphorylation that was inhibited by VTKI. Of note, a post-radiation therapy induction of phospho-p42/p44 was observed on HMME7 cells, and this effect was inhibited by a pretreatment with gefitinib. Based on combination indexes (Chou and Talalay analyses), the associations gefitinib-radiation therapy, VTKI-radiation therapy, VTKI-gefitinib, and gefitinib-VTKI-radiation therapy were found to be additive, slightly synergistic, and markedly synergistic, respectively, for the cytotoxicity on HMME7 cells. Among molecular explanatory factors that were examined, the combination gefitinib-radiation therapy totally abolishes DNA-dependent protein kinase expression, and gefitinib attenuates the radiation therapy-induced enhancement of ERCC1 and augments the VTKI-induced CD95 enhancement. The existence of a radiation therapy-dependent neoangiogenesis may be related to the induction of EGFR pathway in endothelial cells, a phenomenon that can be attenuated by anti-EGFR drugs like gefitinib. In complement to the direct antitumor effects of radiation therapy and anti-EGFR drugs, a strong antiangiogenic effect may be obtained with therapeutic strategies combining radiation therapy with EGFR and VEGFR-2 targeting agents.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Enzyme Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Blotting, Western , Cell Division/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Epidermal Growth Factor/physiology , Gefitinib , Humans , Quinazolines/pharmacology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/physiology
18.
Eur J Cancer ; 41(10): 1467-73, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15919200

ABSTRACT

New drugs and new combinations of drugs have recently shown promising clinical activity in hormone refractory prostate cancer. We studied the association of gefitinib with trastuzumab on the androgen-refractory prostate cancer cell line DU145 expressing both epidermal growth factor receptor (EGFR) and HER-2. Drug combinations with radiotherapy (RT) were considered along with the analysis of factors linked to cell proliferation and apoptosis. The antitumour effects of gefitinib were more pronounced than those observed with trastuzumab. In mice receiving the gefitinib-trastuzumab combination, reduction in tumour volume was inferior to that predicted by the observed impact of the agents alone. The presence of trastuzumab markedly attenuated the relative increase on p27 expression and the Bax:Bcl2 ratio induced by gefitinib. The combination gefitinib-RT had similar antitumour effects as those predicted by the impact of the individual treatments, whereas the effect of the trastuzumab-RT combination was inferior to that predicted by the individual effects. The present data should be borne in mind when designing new clinical schedules for treatment of hormone-refractory prostate cancer including the use of HER inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Gefitinib , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Quinazolines/administration & dosage , Trastuzumab , Xenograft Model Antitumor Assays
19.
Anticancer Drugs ; 16(3): 309-16, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15711183

ABSTRACT

Single-agent docetaxel (Taxotere) treatment has recently demonstrated promising clinical activity in patients with advanced hormone-refractory prostate cancer. Taxanes were recently found to upregulate the tumoral activity of thymidine phosphorylase (TP), a key cellular enzyme [transformation of 5'-deoxy-5-fluorouridine (5'-DFUR) into 5-fluorouracil] in the activation cascade of capecitabine (Xeloda). We tested (cytotoxic effects and molecular mechanisms) the Taxotere-5'-DFUR combination on hormone-refractory prostate cancer cell lines (DU145 and PC3). Cells were exposed to Taxotere and/or 5'-DFUR in three different sequences: Taxotere was given alone for 48 h, then 5'-DFUR was added for 48 h; Taxotere and 5'-DFUR together during 96 h or 5'-DFUR was given alone for 48 h then Taxotere was added for 48 h. The drug sequence Taxotere applied first followed by 5'-DFUR led to synergistic cytotoxic effects on both cell lines; the other sequences resulted in simple additivity. Taxotere did not modify TP activity while it decreased thymidylate synthase activity. There was an increase in CD95 cellular membrane levels following exposure to Taxotere-5'-DFUR, which is in agreement with the supra-additive cytotoxic combination. This observation may serve as a preclinical rationale for a next step testing the Taxotere-capecitabine combination at the clinical level in prostate cancer patients.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Enzyme Inhibitors/pharmacology , Floxuridine/therapeutic use , Prostatic Neoplasms/drug therapy , Taxoids/therapeutic use , Thymidine Phosphorylase/metabolism , Thymidylate Synthase/antagonists & inhibitors , Cell Line, Tumor , Docetaxel , Drug Screening Assays, Antitumor , Drug Synergism , Drug Therapy, Combination , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Thymidylate Synthase/metabolism
20.
Eur J Cancer ; 40(18): 2837-44, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15571968

ABSTRACT

Epidermal growth factor receptor (EGFR) and HER-2 are associated with a poor prognosis in various cancers, including prostate cancer. Inhibition of these receptors may provide a treatment for hormone-refractory prostate cancer. The presence of HER-2 (Western blot) and EGFR (5830 fmol/mg protein, ligand-binding assay) was assessed in the hormone-refractory human prostate cancer cell line, DU-145. Cells were exposed to the selective EGFR-TKI (EGFR tyrosine kinase inhibitor) gefitinib ('Iressa; ZD1839) and/or the HER-2-targeted monoclonal antibody trastuzumab ('Herceptin'), for 96 h. Irradiation (RX) at 6 Gy the dose causing 50% growth inhibition, was applied 48 h after the start of drug treatment. There was a dose-related effect on cell survival for both ZD1839 and trastuzumab treatments. Combining ZD1839 and trastuzumab led to less than additive effects on cell survival. Chou and Talalay representations further characterised this less than additive effect on cell survival. The application of ZD1839 led to a marked elevation in the level of the negative regulator of cell division, p27. The ZD1839-trastuzumab combination had less of an impact on p27 expression compared with the effect of ZD1839 treatment alone. The lowest expression of the apoptotic-related protein, Bax, was observed in the presence of the drug combination. There was a significant interaction (synergism) between RX and either ZD1839 or trastuzumab treatments. In contrast, the drug combination with RX resulted in antagonistic cytotoxic effects. These results indicate an antagonistic interaction between EGFR and HER-2 targeting and provide molecular mechanisms supporting this observation. Data from DU-145 cells suggest that dual targeting of EGFR and HER-2 may be inappropriate for the treatment of hormone-refractory prostate cancer, especially in the context of their combination with RX.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Humans , Male , Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...