Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Angiogenesis ; 12(1): 69-79, 2009.
Article in English | MEDLINE | ID: mdl-19212818

ABSTRACT

Mature circulating endothelial cell (CEC) as well as endothelial progenitor populations may reflect the activity of anti-angiogenic agents on tumor neovasculature or even constitute a target for anti-angiogenic therapy. We investigated the behavior of CECs in parallel with hematopoietic progenitor cells (HPCs) in the blood of renal cell cancer patients during sunitinib treatment. We analyzed the kinetics of a specific population of small VEGFR2-expressing CECs (CD45(neg)/CD34(bright)), HPCs (CD45(dim)/CD34(bright)), and monocytes in the blood of 24 renal cell cancer (RCC) patients receiving 50 mg/day of the multitargeted VEGF inhibitor sunitinib, on a 4-week-on/2-week-off schedule. Blood was taken before treatment (C1D1), on C1D14, C1D28, and on C2D1 before the start of cycle 2. Also plasma VEGF and erythropoietin (EPO) were determined. Remarkably, while CD34(bright) HPCs and monocytes decreased during treatment, CD34(bright) CECs increased from 69 cells/ml (C1D1) to 180 cells/ml (C1D14; P = 0.001) and remained high on C1D28. All cell populations recovered to near pre-treatment levels on C2D1. Plasma VEGF and EPO levels were increased on C1D14 and partly normalized to pre-treatment levels on C2D1. In conclusion, opposite kinetics of two circulating CD34(bright) cell populations, HPCs and small CECs, were observed in sunitinib-treated RCC patients. The increase in CECs is likely caused by sunitinib targeting of immature tumor vessels.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Endothelial Cells/cytology , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Pyrroles/therapeutic use , Adult , Aged , Aged, 80 and over , Antigens, CD34/metabolism , Biomarkers, Tumor/blood , Blood Cell Count , Carcinoma, Renal Cell/blood , Endothelial Cells/drug effects , Erythropoietin/blood , Female , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Humans , Indoles/pharmacology , Kidney Neoplasms/blood , Kinetics , Male , Middle Aged , Pyrroles/pharmacology , Sunitinib , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/blood
2.
Clin Cancer Res ; 14(18): 5884-92, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794101

ABSTRACT

PURPOSE: A disturbed myeloid lineage development with abnormally abundant neutrophils and impaired dendritic cell (DC) differentiation may contribute to tumor immune escape. We investigated the effect of sunitinib, a tyrosine kinase inhibitor of fms-like tyrosine kinase-3, KIT, and vascular endothelial growth factor receptors, on myeloid differentiation in renal cell cancer (RCC) patients. EXPERIMENTAL DESIGN: Twenty-six advanced RCC patients were treated with sunitinib in a 4-week on/2-week off schedule. Enumeration and extensive phenotyping of myeloid subsets in the blood was done at baseline and at weeks 4 and 6 of the first treatment cycle. Baseline patient data were compared with sex- and age-matched healthy donor data. RESULTS: Baseline frequencies of DC subsets were lower in RCC patients than in healthy donors. After 4 weeks of sunitinib treatment, a generalized decrease in myeloid frequencies was observed. Whereas neutrophils and monocytes, which were both abnormally high at baseline, remained low during the 2-week off period, DC rates recovered, resulting in a normalized myeloid lineage distribution. Subsequent to sunitinib treatment, an increase to high levels of myeloid DC (MDC) subset frequencies relative to other myeloid subsets, was specifically observed in patients experiencing tumor regression. Moreover, high CD1c/BDCA-1(+) MDC frequencies were predictive for tumor regression and improved progression-free survival. CONCLUSION: The sunitinib-induced myeloid lineage redistribution observed in advanced RCC patients is consistent with an improved immune status. Immunologic recovery may contribute to clinical efficacy as suggested by the finding of highly increased MDC frequencies relative to other myeloid subsets in patients with tumor regression.


Subject(s)
Carcinoma, Renal Cell/immunology , Dendritic Cells/immunology , Indoles/pharmacology , Kidney Neoplasms/immunology , Myeloid Cells/immunology , Pyrroles/pharmacology , Adult , Aged , Aged, 80 and over , Antigens, CD1/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cell Lineage , Disease-Free Survival , Female , Glycoproteins/metabolism , Humans , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Male , Middle Aged , Pyrroles/therapeutic use , Sunitinib
3.
Eur J Cancer ; 43(2): 433-42, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17097285

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) and cyclo-oxygenase (COX) inhibitors are anti-inflammatory agents that have also shown to be useful in anticancer therapy. In the present study, we show that the specific COX-2 inhibitor celecoxib enhances the inhibitory effect of doxorubicin (dox) on human MDA-MB231 breast tumour growth in vivo and in vitro. We also found that celecoxib increased the intracellular accumulation and retention of dox in vitro. Since the NSAID indomethacin and the specific COX-2 inhibitor NS398 did not affect the in vitro actions of dox, these effects are likely to be mediated via a COX-independent mechanism. It has been suggested that some COX-inhibitors can enhance the actions of cytostatics by overcoming multidrug resistance through the inhibition of ABC-transporter proteins. However, we found that the three main ATP-binding cassette (ABC)-transporter proteins, implicated in dox transport, were inactive in MDA-MB231 cells. Therefore, the finding that the P-glycoprotein (P-gp) blocker PSC833 also increased cellular accumulation of dox was unexpected. In order to unravel the molecular mechanisms involved in dox accumulation, we examined the involvement of NF-kappaB, as this transcription factor has been implicated in celecoxib action as well as in chemoresistance. We found that celecoxib and PSC833, but not indomethacin or NS398, almost completely inhibited basal- and dox induced NF-kappaB gene-reporter activity and p65 subunit nuclear translocation. Furthermore, the NF-kappaB inhibitor PDTC mimicked the actions of celecoxib and PSC833 on cell growth and on intracellular accumulation of dox, suggesting that NF-kappaB is functionally involved in the actions of these compounds. In conclusion, we show that structurally different compounds, among which are celecoxib and PSC833, increase the intracellular accumulation of dox and enhance dox induced cytotoxicity in MDA-MB231 breast cancer cells most likely via the modulation of NF-kappaB activity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , NF-kappa B/metabolism , Pyrazoles/pharmacology , Sulfonamides/pharmacology , ATP-Binding Cassette Transporters/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Celecoxib , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Doxorubicin/pharmacokinetics , Female , Humans , Mice , Mice, Inbred BALB C
4.
Thromb Haemost ; 98(2): 440-50, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17721629

ABSTRACT

Circulating cells of several lineages are thought to participate in angiogenesis and tumor growth. Experimental studies in tumor-bearing mice have pointed to the potential importance of VEGF-responding circulating (endothelial) progenitor cells in tumor growth. We have studied circulating CD31- and/or CD34-positive cell populations with a low to moderate VEGFR2 expression in human volunteers and cancer patients. We recognized four cell populations, which were further characterized by their content of major hematopoietic progenitor, monocytic, endothelial and platelet markers. After establishing the test-retest stability of the measurements in nine patients, we determined the frequencies of the various cell populations in a group of 20 volunteers and 14 cancer patients. Two populations were markedly increased in cancer patients. Small CD45(neg)/CD34(bright)/VEGFR2+ cells amounted to 12 and 64 cells/ml (P < 0.0001), respectively, and 246/ml and 578/ml VEGFR2+/CD45(bright) (/CD14+) monocytic cells were present in controls and cancer patients, respectively (P = 0.017). A third population of CD45(dim)/CD34(bright)/VEGFR2(low) cells amounted to 25 and 30 cells/ml (P = 0.38). Unexpectedly, a population of mainly anucleated CD45(low)/CD31(bright)/CD41(bright) cells was present in numbers of 9,076 and 16,697/ml (P = 0.04) in volunteers and cancer patients, which contained a VEGFR2(low) (compared to IgG isotype control) expressing population amounting to 1,142 and 1,642 cells/ml (P = 0.12). This fourth population probably reflects large platelets. The role of the herein identified VEGFR2+ circulating cell populations deserve further investigation in cancer patients treated with VEGF(R)-targeted therapies. Quantification of such cell populations in the blood of tumor patients may be valuable to monitor the efficacy of anti-angiogenic treatment.


Subject(s)
Endothelial Cells/cytology , Neoplasms/blood , Stem Cells/chemistry , Vascular Endothelial Growth Factor Receptor-2/analysis , Antigens, CD34/analysis , Blood Cells/cytology , Blood Circulation , Case-Control Studies , Endothelial Cells/chemistry , Flow Cytometry , Humans , Leukocyte Common Antigens/analysis , Stem Cells/cytology
5.
Cancer Lett ; 241(2): 309-19, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16337338

ABSTRACT

Tumors generally display a high glycolytic rate. One consequence of increased glycolysis is the non-enzymatic glycation of proteins leading to the formation of advanced glycation end-products (AGEs). Therefore, we studied the presence of AGEs in non-small cell lung cancer and consequences thereof. We show the presence of two AGEs, i.e. the major AGE N(epsilon)-(carboxymethyl)lysine (CML) and the methylglyoxal-arginine adduct argpyrimidine, in human non-small cell lung cancer tissues by immunohistochemistry. We found in squamous cell carcinoma and adenocarcinoma tissues a strong CML positivity in both tumour cells and tumour-surrounding stroma. In contrast, argpyrimidine positivity was predominantly found in tumor cells and was strong in squamous cell carcinomas, but only weak in adenocarcinomas (2.6+/-0.5 vs. 1.2+/-0.4, respectively; P<0.005). In accordance, argpyrimidine was found in the human lung squamous carcinoma cell line SW1573, while it was almost absent in the adenocarcinoma cell line H460. Heat shock protein 27 (Hsp27) was identified as a major argpyrimidine-modified protein. In agreement with a previously described anti-apoptotic activity of argpyrimidine-modified Hsp27, the percentage of active caspase-3 positive tumor cells in squamous cell carcinomas was significantly lower when compared to adenocarcinomas. In addition, incubation with cisplatin induced almost no caspase-3 activation in SW1573 cells while a strong activation was seen in H460 cells; which was significantly reduced by incubation with an inhibitor of glyoxalase I, the enzyme that catalyzes the conversion of methylglyoxal. These findings suggest that a high level of argpyrimidine-modified Hsp27 is a mechanism of cancer cells for evasion of apoptosis.


Subject(s)
Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung , Glycation End Products, Advanced/metabolism , Heat-Shock Proteins/metabolism , Ornithine/analogs & derivatives , Pyrimidines/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Caspase 3 , Caspases/metabolism , Enzyme-Linked Immunosorbent Assay , Glucose/metabolism , Glycolysis , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Ornithine/pharmacology , Tumor Cells, Cultured
6.
Clin Cancer Res ; 11(17): 6240-6, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16144927

ABSTRACT

PURPOSE: SU6668 is a tyrosine kinase inhibitor which targets platelet-derived growth factor receptor-beta, fibroblast growth factor receptor-1, vascular endothelial growth factor receptor-2, and KIT. We did a phase I study to define the maximum tolerated dose and to assess the pharmacokinetics of SU6668 administered orally thrice daily with food. PATIENTS AND METHODS: Patients with histologically proven, advanced, and progressive solid tumors were included at a starting dose level of 400 mg/m2 thrice daily. The early onset of dose-limiting toxicities (DLT) required dose reductions to 100 and 200 mg/m2 thrice daily. Pharmacokinetics was done on days 1, 28, and 56. RESULTS: Sixteen patients were included. Two of the first three patients developed DLTs, which consisted of grade 4 fatigue and grade 3 serositis-like pains. Six patients at dose level 100 mg/m2 thrice daily experienced no DLT. At dose level 200 mg/m2 thrice daily, two out of seven patients experienced DLTs consisting of grade 3 abdominal pain, grade 4 anorexia and grade 3 nausea/vomiting. Increasing doses resulted in a disproportional increase in area under the curve and C(max) (peak plasma concentration). Both variables, however, decreased significantly on days 28 and 56 compared with day 1 (P < 0.05). No objective responses were observed. Acute phase response, probably mediated by interleukin-6, was observed in serial blood samples. CONCLUSIONS: The maximum tolerated dose of SU6668 given orally, thrice daily under fed conditions, is 100 mg/m2. Because of the low plasma levels reached at this dose level, the efficacy of SU6668 as a single agent is not to be expected.


Subject(s)
Indoles/administration & dosage , Neoplasms/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Drug Administration Schedule , Female , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Oxindoles , Propionates , Pyrroles/adverse effects , Pyrroles/pharmacokinetics
7.
Ann N Y Acad Sci ; 1043: 725-33, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16037299

ABSTRACT

Tumors are generally characterized by an increased glucose uptake and a high rate of glycolysis. Since one consequence of an elevated glycolysis is the nonenzymatic glycation of proteins, we studied the presence of advanced glycation end products (AGEs) in human cancer tissues. We detected the presence of the AGEs N(epsilon)-(carboxymethyl)lysine (CML) and argpyrimidine in several human tumors using specific antibodies. Because AGEs have been associated with the etiology of a variety of different diseases, these results suggest that CML and argpyrimidine could be implicated in the biology of human cancer.


Subject(s)
Glycation End Products, Advanced/analysis , Lysine/analogs & derivatives , Neoplasms/chemistry , Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Glycolysis , Humans , Lysine/analysis , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Pyruvaldehyde/analysis
8.
Clin Cancer Res ; 10(3): 1180-91, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871998

ABSTRACT

PURPOSE: Expression of aminopeptidase N/CD13 can be detected in several solid tumor types. Thus far, the role of CD13 in ovarian cancer has not been studied. We have investigated the expression pattern and biological function of CD13 in ovarian cancer. EXPERIMENTAL DESIGN: First, we studied the expression of CD13 in ovarian cancer tissue of 15 patients representing three different histological types (5 patients each) by immunohistochemistry. We then stably transfected the IGROV-1 human ovarian cancer cell line with a CD13 expression vector and examined the biological effect of CD13 in vitro and in vivo. RESULTS: The expression of CD13 in ovarian cancer was associated with the histological subtype: CD13 expression in tumor cells was observed in 80-100% of the patients with a serous or mucinous carcinoma and in only 20% of the clear cell carcinoma patients. In all patients' tumor samples, CD13-positive blood vessels were present. CD13 overexpression in IGROV-1 cells did not affect in vitro cell growth and sensitivity to doxorubicin, cisplatin, or gemcitabine. CD13 overexpression reduced invasion in Matrigel, which appeared to be independent of the aminopeptidase activity of CD13. Furthermore, the growth rate of IGROV-1/CD13 xenografts was reduced. The area of the vessel lumens was enlarged in a small percentage of vessels in the CD13-overexpressing xenografts. In addition, the CD13-overexpressing tumors were less sensitive to cisplatin. CONCLUSIONS: CD13 is expressed in tumor as well as endothelial cells in human ovarian cancer. Our results suggest that CD13 overexpression affects ovarian cancer growth, vascular architecture, and response to chemotherapy. Further elucidation of the mechanism of the observed effects of CD13 is warranted to better understand its role in the pathophysiology of ovarian cancer.


Subject(s)
CD13 Antigens/biosynthesis , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Mucinous/metabolism , Aminopeptidases/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Division , Cell Line, Tumor , Cell Membrane/metabolism , Collagen/pharmacology , Cystadenocarcinoma, Serous/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Female , Humans , Immunohistochemistry , Laminin/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Plasmids/metabolism , Proteoglycans/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Time Factors , Transfection
9.
Clin Cancer Res ; 8(12): 3747-54, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12473585

ABSTRACT

PURPOSE: On the basis of the finding of marked overexpression in angiogenic microvessels, aminopeptidase N/CD13 has recently been suggested to play a prominent role in tumor angiogenesis. A soluble form of CD13 (sCD13) is present in human plasma, but its role in cancer has not been addressed. We hypothesized that sCD13 would be shed by tumor cells and/or endothelial cells lining tumor vessels, giving high levels of sCD13 in intratumoral fluid (TF) deposits and in malignant effusions. If so, sCD13 could be a convenient potential marker for tumor load and/or activated tumor endothelium. EXPERIMENTAL DESIGN: We have measured the specific sCD13 activity in effusions from 90 cancer patients and 12 patients with a nonmalignant condition, and studied its relationship with other major (anti-)angiogenic factors. In a separate group of patients (n = 41), the relationship of sCD13 activity in plasma with tumor load was studied. RESULTS: The sCD13 activity was highest in plasma from cancer patients 71.9 (fmol/ml/s hydrolyzed substrate) versus 42.4 for healthy subjects. In TF, malignant effusions, and nonmalignant effusions, the activities were 52.8, 33.5, and 18.6, respectively. We further studied the relationship of sCD13 with tumor load as well as with vascular endothelial growth factor (VEGF), endostatin, matrix metalloproteinase (MMP)-2, MMP-9, urokinase-type plasminogen activator, and plasmin. A significant correlation of sCD13 activity in plasma was found with tumor load (r = 0.68; P = 0.01), suggesting that plasma sCD13 is, at least, partly originating from tumor(-endothelium). The concentrations of VEGF and endostatin and the activities of urokinase-type plasminogen activator and MMP-9, but not MMP-2, were significantly higher in TF compared with all other effusions. In TF, a correlation between sCD13 and VEGF was found (r = 0.67; P = 0.03). No correlation of sCD13 with the other protease activities was found. CONCLUSION: The sCD13 activity is elevated in plasma and effusions of cancer patients. A strong correlation of plasma sCD13 with tumor load was found. On the basis of these results, the potential of sCD13 activity as a tumor and/or angiogenesis marker warrants further investigation.


Subject(s)
Ascitic Fluid/metabolism , Biomarkers, Tumor/metabolism , CD13 Antigens/metabolism , Neoplasms/metabolism , Pleural Effusion/metabolism , Adult , Collagen/metabolism , Endostatins , Endothelial Growth Factors/metabolism , Female , Fibrinolysin/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Immunoblotting , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , Neoplasms/pathology , Peptide Fragments/metabolism , Pleural Effusion/pathology , Prognosis , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Clin Cancer Res ; 9(5): 1648-55, 2003 May.
Article in English | MEDLINE | ID: mdl-12738717

ABSTRACT

INTRODUCTION: Vascular endothelial growth factor (VEGF) is a key regulator in angiogenesis. Preclinical and clinical data support the role of VEGF and angiogenesis in renal cell carcinoma (RCC), melanoma (M), and soft tissue sarcoma (STS). The tyrosine kinase inhibitor SU5416 is a potent inhibitor of the VEGF receptors 1 and 2. EXPERIMENTAL DESIGN: We investigated 145 mg/m(2) SU5416 twice weekly in patients with advanced or metastatic RCC, M, and STS. The primary objectives were efficacy and safety. Disease assessments were performed after 4 and 8 weeks of treatment and every 2 months thereafter. Documented stable disease (SD) lasting for > or =3 months was considered an antitumor response. RESULTS: A group of 29 patients was entered in the RCC trial, 20 patients in the M trial, and 31 patients in the STS trial. Response was observed in 6 (1 minor response and 5 SDs) of 24 evaluable patients (25%) in the RCC group, 6 (1 minor response and 5 SDs) of 26 patients (23%) in the STS group, and none of the patients in the M group. Progression-free survival ranged from 7 to 252 days (median 59 days) in the RCC group, from 7 to 260 days (median 60 days) in the STS group, and from 14 to 139 days (median 41 days) in the M group. Toxicities observed were those reported previously for SU5416. CONCLUSION: SU5416 single agent is well tolerated. The antitumor response was low in patients with RCC and STS, whereas no responses were seen in patients with M.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Enzyme Inhibitors/therapeutic use , Indoles/therapeutic use , Pyrroles/therapeutic use , Adult , Aged , Carcinoma, Renal Cell/drug therapy , Disease-Free Survival , Female , Humans , Kidney Neoplasms/drug therapy , Male , Melanoma/drug therapy , Middle Aged , Neovascularization, Pathologic , Protein-Tyrosine Kinases/antagonists & inhibitors , Sarcoma/drug therapy , Sarcoma/metabolism , Skin Neoplasms/drug therapy , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL