Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Res ; 65(3): 767-79, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15705873

ABSTRACT

Breast cancer is a heterogeneous disease whose evolution is difficult to predict by using classic histoclinical prognostic factors. Prognostic classification can benefit from molecular analyses such as large-scale expression profiling. Using immunohistochemistry on tissue microarrays, we have monitored the expression of 26 selected proteins in more than 1,600 cancer samples from 552 consecutive patients with early breast cancer. Both an unsupervised approach and a new supervised method were used to analyze these profiles. Hierarchical clustering identified relevant clusters of coexpressed proteins and clusters of tumors. We delineated protein clusters associated with the estrogen receptor and with proliferation. Tumor clusters correlated with several histoclinical features of samples, including 5-year metastasis-free survival (MFS), and with the recently proposed pathophysiologic taxonomy of disease. The supervised method identified a set of 21 proteins whose combined expression significantly correlated to MFS in a learning set of 368 patients (P < 0.0001) and in a validation set of 184 patients (P < 0.0001). Among the 552 patients, the 5-year MFS was 90% for patients classified in the "good-prognosis class" and 61% for those classified in the "poor-prognosis class" (P < 0.0001). This difference remained significant when the molecular grouping was applied according to lymph node or estrogen receptor status, as well as the type of adjuvant systemic therapy. In multivariate analysis, the 21-protein set was the strongest independent predictor of clinical outcome. These results show that protein expression profiling may be a clinically useful approach to assess breast cancer heterogeneity and prognosis in stage I, II, or III disease.


Subject(s)
Breast Neoplasms/classification , Breast Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Female , Humans , Immunohistochemistry , Middle Aged , Multivariate Analysis , Prognosis , Protein Array Analysis , Proteomics , Reproducibility of Results
2.
Clin Cancer Res ; 11(2 Pt 1): 712-9, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15701860

ABSTRACT

PURPOSE: To test the efficacy of selective therapy against cyclooxygenase-2 in combination with a low-dose regimen of a cytotoxic agent in the treatment of juvenile hematopoietic malignancies in the experimental model, Friend disease. EXPERIMENTAL DESIGN: Juvenile erythroleukemic mice (n = 8) received no treatment, celecoxib (1600 mg/kg/d), vinblastine (0.5 microg/g twice weekly), vehicle controls, or celecoxib + vinblastine combination (n = 9) over a 6-month period from time of tumor induction. Overt toxicity was assessed daily and recorded weekly. RESULTS: Among randomly selected mice from celecoxib treatment groups, plasma concentrations ranged from 2 to 6 micromol/L. As a single agent, celecoxib was not associated with any apparent toxicity. Monotherapy with vinblastine, however, caused early mortality marked by severe diarrhea, lethargy, and weight loss. At the tested doses, neither vinblastine nor celecoxib enhanced survival as monotherapies. Coadministration of these two drugs alleviated the overt toxicity associated with vinblastine and resulted in a significant increase in survival (P < 0.05). Survivors sampled throughout the study showed a trend to decreased weight loss and hematocrit levels among all groups, but significance was evidenced earlier in the vinblastine monotherapy group overall (P < 0.05). Despite similar degree of splenomegaly, histologic analysis revealed preserved splenic mantle architecture from mice given combination therapy compared with those sampled from mice on all other monotherapies, exhibiting a more diffuse burden of blasts and destruction of germinal centers. CONCLUSION: We propose that addition of a selective cyclooxygenase-2 inhibitor to a modified low-dose conventional chemotherapeutic regimen protects juvenile mice with Friend disease from succumbing to low-dose cytotoxicity, in part, by neutralizing acute inflammatory responses.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Leukemia, Erythroblastic, Acute/drug therapy , Leukemia, Experimental/drug therapy , Prostaglandin-Endoperoxide Synthases/chemistry , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Vinblastine/therapeutic use , Animals , Cardiovascular Diseases/chemically induced , Celecoxib , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Friend murine leukemia virus/genetics , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Inbred BALB C , Prostaglandin-Endoperoxide Synthases/metabolism , Survival Rate , Treatment Outcome
3.
J Cardiovasc Pharmacol ; 45(1): 61-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15613981

ABSTRACT

The inflammation in response to vascular injury is becoming increasingly recognized as a potential contributor to restenosis. Cyclooxygenase-2 (COX-2) is the inducible form of cyclooxygenase and has been shown to be involved in the proinflammatory response of vascular tissue. Bilateral femoral artery lesions were induced by air desiccation in New Zealand White rabbits followed by high cholesterol diet feeding for 28 days. Balloon injury and stent implantation were performed at the preinjured vessel segments. Immunostaining showed that uninjured vessel segments stained positive only for COX-1 but not for COX-2. Injured vessel segments showed, in addition to COX-1, significant positive staining for COX-2. In the efficacy study, celecoxib (75 mg/kg/d) was administered orally beginning 3 hours before balloon injury or stent implantation on day 28 and daily for 21 days. Monocyte chemoattractant protein-1 (MCP-1) and matrix metalloproteinase-2 and -9 (MMPs) expression were quantified in arterial extracts 4 days after balloon injury by Western blot and gelatin zymography. Morphometric analysis and immunostaining for macrophages were performed 21 days after balloon injury. Celecoxib treatment significantly decreased MCP-1 expression (P < 0.01). Neointimal hyperplasia was significantly inhibited by celecoxib in both balloon injury and stent models (0.49 +/- 0.20 versus 0.70 +/- 0.35 mm2 from balloon injury model, P < 0.05, and 0.81 +/- 0.25 versus 1.69 +/- 0.43 mm2 from stent model, P < 0.05), accompanied by reduced macrophage infiltration. We conclude that celecoxib decreases the inflammatory response and intimal hyperplasia following vascular injury, possibly through inhibition of MCP-1 expression, implying a pivotal role of inflammation in the pathogenesis of restenosis.


Subject(s)
Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Chemokine CCL2/antagonists & inhibitors , Cyclooxygenase Inhibitors/pharmacology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Tunica Intima/drug effects , Angioplasty, Balloon , Animals , Arteriosclerosis/etiology , Celecoxib , Chemokine CCL2/biosynthesis , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacokinetics , Female , Hyperplasia , Iliac Artery/drug effects , Iliac Artery/pathology , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors , Pyrazoles/pharmacokinetics , Rabbits , Stents , Sulfonamides/pharmacokinetics , Tunica Intima/pathology
4.
Oncogene ; 23(14): 2564-75, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14743203

ABSTRACT

ERBB2 is a transmembrane tyrosine kinase receptor encoded by a gene located in chromosome region 17q12. Overexpression of ERBB2, generally by way of gene amplification, plays a role in mammary oncogenesis. This alteration can be overcome by use of the humanized monoclonal antibody trastuzumab (Herceptin). Accurate determination of ERBB2 status is required for appropriate use of this targeted therapy and is currently analysed by immunohistochemistry (IHC) on tissue sections and/or fluorescence in situ hybridisation (FISH) on interphase chromosomes. We have studied the gene expression profiles of a series of 213 breast tumours and 16 breast cancer cell lines with known ERBB2 status, using Ipsogen's DiscoveryChip microarrays with approximately 9000 cDNAs. We have identified 36 genes and expressed sequence tags that were differentially expressed in tumours and in cell lines with and without ERBB2 protein overexpression. This ERBB2-specific gene expression signature (GES) contained 29 overexpressed genes including the ERBB2 gene itself, five genes located in its immediate vicinity on 17q12, non-17q genes such as GATA4 and eight downregulated genes including oestrogen receptor alpha (ER). Some correlations were validated at the protein level using IHC on tissue microarrays. The GES was able to distinguish ERBB2-negative and -positive cancer samples, as well as FISH-negative and FISH-positive ERBB2 2+ IHC samples.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, erbB-2 , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Breast Neoplasms/classification , Cell Line, Tumor , Chromosome Mapping , Chromosomes, Human, Pair 17 , Down-Regulation , Expressed Sequence Tags , Female , Gene Expression Profiling , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Middle Aged , Nucleic Acid Amplification Techniques , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Reproducibility of Results , Sensitivity and Specificity , Trastuzumab
5.
Oncogene ; 23(13): 2305-14, 2004 Mar 25.
Article in English | MEDLINE | ID: mdl-14743207

ABSTRACT

It is well established that selective COX-2 inhibitors exhibit potent effects against progression of select solid tumours. However, their effects on liquid tumours have not been fully established. By taking advantage of murine Friend Disease we have shown a strong antileukemic effect of celecoxib by determining novel in vitro targets. Western blot analyses revealed the expression of COX-2 in a panel of Friend Virus-transformed, splenic-derived primary erythroleukemic blasts and established cell lines generated in our laboratory. We have shown that celecoxib at concentrations as low as 20 microM significantly suppresses proliferation of the selected murine erythroleukemia cell line HB60-5. The greatest proliferative inhibition was seen at 40 microM of celecoxib, resulting in apoptosis. Our results also demonstrate that treatment of the established murine erythroleukemia cell line HB60-5 with celecoxib results in suppression of c-Kit and erythropoietin receptor (Epo-R) phosphorylation resulting in apoptosis, likely through decreased levels of survival factors. However, upon overexpression of c-Kit alone in these cells a significant increase in survival and twofold increase in proliferation in the presence of celecoxib were observed (P < 0.05). Finally, since responsiveness of our murine erythroleukemia cell lines to celecoxib is above the reported physiologically achievable levels in vivo, we have provided in vitro evidence to suggest that reduced sensitivity of erythroleukemic cells to lower doses of celecoxib may be a consequence of the loss of wild-type p53. These findings are pivotal in addressing potential discrepancies associated with sensitivity of murine erythroleukemic cells to celecoxib in vitro versus in vivo.


Subject(s)
Drug Resistance, Neoplasm/physiology , Leukemia, Erythroblastic, Acute/drug therapy , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Erythropoietin/metabolism , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents , Celecoxib , Mice , Phosphorylation , Phosphotransferases/metabolism , Pyrazoles
6.
Cancer Biol Ther ; 2(6): 713-8, 2003.
Article in English | MEDLINE | ID: mdl-14688483

ABSTRACT

Cyclooxygenase 2 (COX-2) is an inducible enzyme involved in the production of prostaglandins and thromboxanes during inflammation. There are now several lines of evidence indicating that increased expression of COX-2 plays a functional role in the development and progression of malignant epithelial cancers. However, there is only limited data regarding the role of COX-2 in melanoma pathogenesis. In the present work, we retrospectively examined lesions through out the development of melanoma and metastatic disease (dysplastic nevi n = 10, melanoma in situ n = 4, stage II melanoma n = 10, stage III n = 4, stage IV n = 3, stage V n = 2, melanoma metastasis lymph nodes n = 13 metastasis to other sites n = 3). COX-2 was consistently observed in keratinocytes, dermal fibroblasts, and inflammatory cells in regions adjacent to benign evi and primary cutaneous melanomas. However, no COX-2 staining was detected in the nevi nor in the primary skin melanoma cells. In addition, COX-2 was undetected in all vertical and radial growth phase cases Interestingly, 13 out of 13 of the lymph node metastasis expressed extremely high levels of COX-2 in overlying epithelium and inflammatory cells, and COX-2 was strongly detected in the metastatic cancer cells per se. For additional information on the expression of COX-2 in malignant melanoma, we determined the expression of COX-2 protein in several different melanoma cell lines. We found that 3We found that 5 out of 7 of the melanoma cells over expressed COX-2 compared to normal melanocytes. Collectively, these data suggest that COX-2 may play a functional role in metastases of melanoma, and treatment with COX-2 inhibitors may be efficacious for malignant melanoma.


Subject(s)
Isoenzymes/metabolism , Melanoma/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Adult , Aged , Arachidonic Acid/pharmacology , Blotting, Western , Cell Line , Cell Line, Tumor , Chromatography, High Pressure Liquid , Culture Media, Serum-Free , Cyclooxygenase 2 , Disease Progression , Female , Fibroblasts/enzymology , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Immunohistochemistry , Isoenzymes/analysis , Lipoxygenase/metabolism , Lymph Nodes/pathology , Male , Mass Spectrometry , Melanocytes/enzymology , Membrane Proteins , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prostaglandin-Endoperoxide Synthases/analysis
7.
Mol Carcinog ; 38(1): 33-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12949841

ABSTRACT

Over a million nonmelanoma skin cancer cases will be reported in the United States this year alone. Currently the primary form of treatment for these types of skin tumors is excision. However, excision of the initial lesion may not be curative because almost 50% of patients with one nonmelanoma skin cancer lesion develop another tumor within the next 5 yr at the site or adjacent to the site of excision. As with other types of epithelial based cancers, there is mounting evidence for the role of cyclooxygenase-2 (COX-2) and its products, particularly prostaglandin E(2) (PGE(2)), in the development of nonmelanoma skin cancer. To avoid the excision process, the present study was designed to evaluate the possible chemotherapeutic effect of directly treating established tumors with a topical formulation of the specific COX-2 inhibitor celecoxib. Skh/hr hairless mice were irradiated three times per wk for 16 wk to induce tumor formation. The mice were then divided into two groups and treated topically with either 500 microg celecoxib or the vehicle for 6 wk. Our results demonstrated that although topical treatment with celecoxib was not able to induce regression of established tumors, it did prevent new tumor formation after the onset of photocarcinogenesis. Although further studies are warranted, these data suggest that topical celecoxib treatment may prove to be effective in preventing the recurrence of tumors at the site of nonmelanoma skin cancer excision.


Subject(s)
Neoplasms, Radiation-Induced/drug therapy , Skin Neoplasms/drug therapy , Sulfonamides/pharmacology , Administration, Topical , Animals , Celecoxib , Cell Division/drug effects , Cyclooxygenase 2 , Dinoprostone/metabolism , Disease Models, Animal , Epidermis/drug effects , Female , Isoenzymes/metabolism , Isoenzymes/radiation effects , Mice , Neoplasms, Radiation-Induced/prevention & control , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandin-Endoperoxide Synthases/radiation effects , Pyrazoles , Skin Neoplasms/etiology , Skin Neoplasms/prevention & control , Sulfonamides/administration & dosage , Ultraviolet Rays
8.
Mol Carcinog ; 38(2): 49-58, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14502644

ABSTRACT

Inflammation, which includes the release of growth factors, proinflammatory cytokines and prostaglandins, the infiltration and activation of inflammatory cells, and the induction of oxidative DNA damage, is known to play a role in cancer development. The combination of damage to the skin resulting from chronic ultraviolet light B (UVB) exposure itself and the inflammatory response it induces is a major source of skin cancer development. Cyclooxygenase-2 (COX-2), an inflammatory enzyme responsible for the production of prostaglandins, is now implicated in the development of epithelial cancers, including squamous cell carcinoma in the skin. Previous work conducted in our laboratory has shown that topical treatment with celecoxib following UVB irradiation inhibits several parameters of acute inflammation, including vascular permeability, the infiltration and activation of neutrophils, and the production of prostaglandin E(2) (PGE(2)). The present studies expanded these observations, demonstrating the ability of topical celecoxib to inhibit acute oxidative damage. In addition, long-term studies illustrate the effectiveness of topical treatment with this drug in reducing chronic inflammation and UVB-induced papilloma/carcinoma formation. This data provides compelling evidence to explore the clinical efficacy of topically applied COX-2 inhibitors for the prevention of human skin cancers.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Dermatitis/prevention & control , Neoplasms, Radiation-Induced/prevention & control , Sulfonamides/pharmacology , Ultraviolet Rays/adverse effects , Administration, Topical , Animals , Carcinoma, Basal Cell , Celecoxib , Cell Division/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , DNA Damage/drug effects , Female , Isoenzymes/antagonists & inhibitors , Mice , Mice, Hairless , Prostaglandin-Endoperoxide Synthases , Pyrazoles , Skin Neoplasms/prevention & control , Sulfonamides/administration & dosage , Tumor Suppressor Protein p53/metabolism
10.
Arthritis Rheum ; 46(7): 1789-803, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12124863

ABSTRACT

OBJECTIVE: To examine cyclooxygenase-2 (COX-2) enzyme expression, its regulation by interleukin-1 beta (IL-1 beta), and the role of prostaglandin E(2) (PGE(2)) in proteoglycan degradation in human osteoarthritic (OA) cartilage. METHODS: Samples of human OA articular cartilage, meniscus, synovial membrane, and osteophytic fibrocartilage were obtained at knee arthroplasty and cultured ex vivo with or without IL-1 beta and COX inhibitors. COX expression was evaluated by immunohistochemistry and Western blot analysis. The enzymatic activity of COX was measured by conversion of arachidonic acid to PGE(2). Cartilage degradation was evaluated by measuring the accumulation of sulfated glycosaminoglycans in the medium. RESULTS: IL-1 beta induced robust expression of COX-2 and PGE(2) in OA meniscus, synovial membrane, and osteophytic fibrocartilage explants, whereas low levels were produced in OA articular cartilage. IL-1 beta also induced cartilage proteoglycan degradation in OA synovial membrane-cartilage cocultures. Increased proteoglycan degradation corresponded to the induction of COX-2 protein expression in, and PGE(2) production from, the synovial membrane. Dexamethasone, neutralizing IL-1 beta antibody, or the selective COX-2 inhibitor, SC-236, attenuated both the IL-1 beta-induced PGE(2) production and cartilage proteoglycan degradation in these cocultures. The addition of PGE(2) reversed the inhibition of proteoglycan degradation caused by SC-236. CONCLUSION: IL-1 beta-induced production of COX-2 protein and PGE(2) was low in OA articular cartilage compared with that in the other OA tissues examined. IL-1 beta-mediated degradation of cartilage proteoglycans in OA synovial membrane-cartilage cocultures was blocked by the selective COX-2 inhibitor, SC-236, and the effect of SC-236 was reversed by the addition of exogenous PGE(2). Our data suggest that induction of synovial COX-2-produced PGE(2) is one mechanism by which IL-1 beta modulates cartilage proteoglycan degradation in OA.


Subject(s)
Cartilage/metabolism , Dinoprostone/physiology , Isoenzymes/metabolism , Osteoarthritis/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Proteoglycans/metabolism , Aged , Antibodies/pharmacology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dexamethasone/pharmacology , Female , Humans , Immunohistochemistry , In Vitro Techniques , Interleukin-1/immunology , Interleukin-1/pharmacology , Male , Membrane Proteins , Menisci, Tibial/metabolism , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Synovial Membrane/metabolism
11.
Cancer Control ; 9(2 Suppl): 28-35, 2002.
Article in English | MEDLINE | ID: mdl-11965228

ABSTRACT

In addition to the well-established pathophysiological role that COX-2 plays in inflammation, recent evidence implies that this isoform may also be involved in multiple biologic events throughout the tumorigenic process. Many epidemiological studies demonstrate that nonsteroidal anti-inflammatory drugs (NSAIDs) reduce the risk of a wide range of tumors. Further, COX-2 is chronically overexpressed in many premalignant, malignant, and metastatic human cancers, and levels of overexpression have been shown to significantly correlate to invasiveness, prognosis, and survival in some cancers. Pharmacological studies consistently demonstrate that COX-2 inhibitors dose-dependently inhibit tumor growth and metastasis in various relevant animal models of cancer. Importantly, several investigators have also shown COX-2 inhibitors may act additively or synergistically with currently used cytotoxics and molecularly targeted agents. Here we present a broad overview of the growing evidence that COX-2 plays a pivotal role throughout oncogenesis and summarize the rationale to explore the use of COX-2 inhibitors for the prevention and/or treatment of cancer as a single agent or in combination with current anticancer modalities.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Celecoxib , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins , Neoplasms/prevention & control , Prognosis , Prostaglandin-Endoperoxide Synthases , Pyrazoles , Receptor, ErbB-2/drug effects
12.
Br J Pharmacol ; 135(4): 891-900, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11861316

ABSTRACT

1. The maintenance of renal function in decompensated cirrhosis is highly dependent on prostaglandins (PGs). Since PG synthesis is mediated by cyclooxygenase-1 and -2 (COX-1 and COX-2), the present study was designed to examine which COX isoform is involved in this phenomenon. 2. Renal COX-1 and COX-2 protein expression and distribution were analysed by Western blot and immunohistochemistry in nine rats with carbon tetrachloride-induced cirrhosis and ascites and 10 control animals. The effects of placebo and selective COX-1 (SC-560) and COX-2 (celecoxib) inhibitors on urine flow (V), urinary excretion of sodium (U(Na)V) and PGE(2) (U(PGE2)V), glomerular filtration rate (GFR), renal plasma flow (RPF), the diuretic and natriuretic responses to furosemide and renal water metabolism were assessed in 88 rats with cirrhosis and ascites. 3. COX-1 protein levels were found to be unchanged in kidneys from cirrhotic rats. In contrast, these animals showed enhanced renal COX-2 protein expression which was focally increased in the corticomedullary region. Although U(PGE2)V was equally reduced by SC-560 and celecoxib, only SC-560 produced a significant decrease in U(Na)V, GFR and RPF and a pronounced impairment in the diuretic and natriuretic responses to furosemide in rats with cirrhosis and ascites. Neither SC-560 nor celecoxib affected renal water metabolism in cirrhotic rats. 4. These results indicate that despite abundant renal COX-2 protein expression, the maintenance of renal function in cirrhotic rats is mainly dependent on COX-1-derived prostaglandins.


Subject(s)
Ascites/metabolism , Ascites/physiopathology , Isoenzymes/metabolism , Kidney/metabolism , Kidney/physiopathology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/physiopathology , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/biosynthesis , Animals , Blotting, Western , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/physiopathology , Celecoxib , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Diuretics/pharmacology , Furosemide/pharmacology , Immunohistochemistry , Male , Membrane Proteins , Pyrazoles/pharmacology , Rats , Rats, Wistar , Sulfonamides/pharmacology , Water/metabolism
13.
Cancer Res ; 62(3): 625-31, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11830509

ABSTRACT

Cyclooxygenase-2 (COX-2) is expressed within neovascular structures that support many human cancers. Inhibition of COX-2 by celecoxib delays tumor growth and metastasis in xenograft tumor models as well as suppresses basic fibroblast growth factor 2 (FGF-2)-induced neovascularization of the rodent cornea. The present studies were undertaken to evaluate possible mechanisms of the antiangiogenic and anticancer effects of celecoxib. Prostaglandin E(2) (PGE(2)) and thromboxane B(2) (TXB(2)) were increased in rat corneas implanted with slow-release pellets containing FGF-2 (338.6 ng of PGE(2)/g and 17.53 ng of TXB(2)/g) compared with normal rat corneas (63.1 ng of PGE(2)/g and 2.0 ng of TXB(2)/g). Celecoxib at 30 mg/kg/day p.o. inhibited angiogenesis (78.6%) and prostaglandin production by 78% for PGE(2) (72.65 ng/g) and 68% for TXB(2) (5.55 ng/g). Decreased prostaglandin production in corneas was associated with a 2.5-fold cellular increase in apoptosis and a 65% decrease in proliferation. Similar reductions in proliferation were observed in neovascular stroma (65-70%) of celecoxib-treated (dietary 160 ppm/day) xenograft tumors as well as in tumor cells (50-75%). Apoptosis was also increased in the tumor cells (2.2-3.0-fold) in response to celecoxib. Thus, the antitumor activity of celecoxib may be attributable, at least in part, to a direct effect on host stromal elements, such as the angiogenic vasculature.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Isoenzymes/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Celecoxib , Cell Division/drug effects , Colonic Neoplasms/blood supply , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cornea/blood supply , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/biosynthesis , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Fibroblast Growth Factor 2/pharmacology , Humans , Isoenzymes/biosynthesis , Membrane Proteins , Mice , Mice, Nude , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Prostaglandin-Endoperoxide Synthases/biosynthesis , Pyrazoles , Rats , Thromboxane B2/biosynthesis , Xenograft Model Antitumor Assays
14.
Adv Exp Med Biol ; 507: 177-84, 2002.
Article in English | MEDLINE | ID: mdl-12664583

ABSTRACT

To understand the potential role of cyclooxygenase (COX) in normal and inflammatory human diseases, we characterized the expression of COX-1 and COX-2 in biopsies of osteoarthritis, atherosclerosis, and cancer. Tissues were prepared for immunohistochemistry by standard methods, and representative cases assayed via Western blot and quantitative RT-PCR. COX-2 was not detected in normal human tissues with few exceptions. Moderate to marked COX-2 was observed in the macula densa (MD) and thick ascending limb (TAL) in human fetal kidneys, but was not detected in neonatal and adult MD and TALs. Low level, constitutive COX-2 was detected in colonic epithelium, peribronchial glands, and pancreatic ductal epithelium. Low to moderate COX-2 was detected basally in the cortex, hippocampus, hypothalamus, and spinal cord, and in reproductive tissues during ovulation, implantation and labor. No COX-2 was detected in the existing vasculature in normal tissues, and was also not expressed throughout the ductus arteriosus. COX-2 was markedly induced in human tissues of osteoarthritis, atherosclerosis and cancer. COX-2 was prominently expressed in the synovium, fibrocartilage of osteophytes, and in the blood vessels in the osteoarthritic (OA) knee joint. COX-2 was also prominently detected in the macrophages/foam cells in atherosclerotic plaques, and in the endothelium overlying and immediately adjacent to the fibrofatty lesion. Moderate- to intense COX-2 expression was consistently observed in the inflammatory cells, neoplastic lesions, and blood vessels in all epithelial-derived human cancers studied. In contrast, COX-1 was relatively ubiquitously observed in both normal and pathophysiological conditions. These data collectively imply COX-2 plays an important role in mediating a variety of inflammatory diseases, and imply COX-2 inhibitors may be effective in the prevention and/or treatment of OA, heart disease, and epithelial cancers.


Subject(s)
Disease , Isoenzymes/metabolism , Lung Neoplasms/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Female , Humans , Lung Neoplasms/pathology , Membrane Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...