Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Mol Cancer Ther ; 5(7): 1817-26, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891468

ABSTRACT

Dysregulation of enzymes involved in prostaglandin biosynthesis plays a critical role in influencing the biological behavior and clinical outcome of several tumors. In human gliomas, overexpression of cyclooxygenase-2 has been linked to increased aggressiveness and poor prognosis. In contrast, the role of prostaglandin E synthase in influencing the biological behavior of human gliomas has not been established. We report that constitutive expression of the microsomal prostaglandin E synthase-1 (mPGES-1) is associated with increased prostaglandin E(2) (PGE(2)) production and stimulation of growth in the human astroglioma cell line U87-MG compared with human primary astrocytes. Consistently, pharmacologic and genetic inhibition of mPGES-1 activity and expression blocked the release of PGE(2) from U87-MG cells and decreased their proliferation. Conversely, exogenous PGE(2) partially overcame the antiproliferative effects of mPGES-1 inhibition and stimulated U87-MG cell proliferation in the absence of mPGES-1 inhibitors. The EP2/EP4 subtype PGE(2) receptors, which are linked to stimulation of adenylate cyclase, were expressed in U87-MG cells to a greater extent than in human astrocytes. PGE(2) increased cyclic AMP levels and stimulated protein kinase A (PKA) activity in U87-MG cells. Treatment with a selective type II PKA inhibitor decreased PGE(2)-induced U87-MG cell proliferation, whereas a selective type I PKA inhibitor had no effect. Taken together, these results are consistent with the hypothesis that mPGES-1 plays a critical role in promoting astroglioma cell growth via PGE(2)-dependent activation of type II PKA.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/metabolism , Glioma/enzymology , Intramolecular Oxidoreductases/metabolism , Astrocytes/drug effects , Astrocytes/enzymology , Cell Proliferation/drug effects , Cyclic AMP-Dependent Protein Kinase Type II , Dinoprostone/pharmacology , Glioma/pathology , Humans , Intramolecular Oxidoreductases/genetics , Microsomes/enzymology , Prostaglandin-E Synthases , Tumor Cells, Cultured
2.
Cancer Res ; 62(20): 5778-84, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12384538

ABSTRACT

Combining anticancer drugs with different mechanisms of action has the potential to enhance antitumor effect. CPT-11 (Camptosar, irinotecan), a topoisomerase I inhibitor, has been shown to be highly effective in the treatment of a variety of cancers. However, its clinical usage is often complicated by late diarrhea. A number of studies have shown that cyclooxygenase (COX)-2 is overexpressed in many forms of human tumors, suggesting that COX-2 inhibition may be useful in the treatment of cancer. In this study, we used two mouse tumor models (HT-29 and colon-26 cells) to evaluate the effect of combining CPT-11 with celecoxib on tumor growth. We also assessed the involvement of COX-2 in the pathogenesis of CPT-11-induced late diarrhea using a rat model. Results indicate that celecoxib enhances the antitumor effect of CPT-11 and reduces the severity of late diarrhea in a dose-dependent manner. The extended benefits of combining celecoxib with CPT-11 may significantly improve the outcome of cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Diarrhea/prevention & control , Isoenzymes/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Camptothecin/administration & dosage , Camptothecin/adverse effects , Celecoxib , Colon/metabolism , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Diarrhea/chemically induced , Dinoprostone/biosynthesis , Dinoprostone/physiology , Drug Administration Schedule , Drug Synergism , HT29 Cells/drug effects , Humans , Irinotecan , Male , Membrane Proteins , Mice , Mice, Inbred BALB C , Mice, Nude , Prostaglandin-Endoperoxide Synthases , Pyrazoles , Rats , Rats, Sprague-Dawley , Sulfonamides/administration & dosage , Weight Loss/drug effects , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 2(1): 1-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12533667

ABSTRACT

Angiogenesis plays a crucial role in tumor development and growth. The present investigation was undertaken to test the potential involvement of the cyclooxygenase-2 (COX-2) pathway in the regulation of angiogenesis and growth in pancreatic cancer. We compared the angiogenic characteristics of a COX-2-positive human pancreatic tumor cell line, BxPC-3, with those of a COX-2-negative pancreatic tumor cell line, AsPC-1. Cultured BxPC-3 cells promoted a marked increase of endothelial cell migration in comparison with migration that occurred in the absence of cancer cells. Furthermore, BxPC-3 cell culture supernatants induced endothelial cell capillary morphogenesis in vitro and neovascularization in vivo. In contrast, cultured AsPC-1 cells elicited a modest effect on endothelial cell migration and neovascularization in vivo. Pretreatment of BxPC-3 cells with the selective COX-2 inhibitor NS-398 (50 micro M) dramatically decreased angiogenic responses of endothelial cells. NS-398 (25-100 micro M) caused inhibition of BxPC-3 cell proliferation but had no effect on AsPC-1 cell growth. SC-560, a selective COX-1 inhibitor, had no effect on growth of either cell lines. These results suggest an involvement of COX-2 in the control of tumor-dependent angiogenesis and growth in certain pancreatic cancers and provide the rational for inhibition of the COX pathway as an effective therapeutic approach for pancreatic tumors.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Isoenzymes/physiology , Neovascularization, Pathologic/enzymology , Nitrobenzenes/pharmacology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Prostaglandin-Endoperoxide Synthases/physiology , Sulfonamides/pharmacology , Capillaries/drug effects , Capillaries/pathology , Cell Division/drug effects , Cell Division/physiology , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/metabolism , Endothelial Growth Factors/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Gene Expression Regulation, Enzymologic/drug effects , Homeostasis , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lymphokines/metabolism , Membrane Proteins , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/enzymology , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Cancer Med ; 4(1): 36-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25296628

ABSTRACT

Severe infusion reactions (SIRs) at rates of 5% or less are known side effects of biological agents, including mAbs such as cetuximab. There are currently no prospectively validated risk factors to aid physicians in identifying patients who may be at risk of experiencing an SIR following administration of any of these drugs. A retrospective analysis of 545 banked serum or plasma samples from cancer patients participating in clinical trials of cetuximab was designed to evaluate whether the presence of pretreatment IgE antibodies against cetuximab, as determined by a commercially available assay system, is associated with SIRs during the initial cetuximab infusion. Patients with a positive test indicating the presence of pretreatment antibodies had a higher risk of experiencing an SIR; however, at the prespecified cutoff utilized in this analysis, the test has a relatively low-positive predictive value (0.577 [0.369-0.766]) and a negative predictive value of 0.961 (0.912-0.987) in an unselected patient population. Data collected in this large retrospective validation study support prior observations of an association between the presence of pretreatment IgE antibodies cross-reactive with cetuximab and SIRs. Further analysis of the test's ability to predict patients at risk of an SIR would be required before this assay could be used reliably in this patient population.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Cross Reactions/immunology , Drug Hypersensitivity/immunology , Immunoglobulin E/immunology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Case-Control Studies , Cetuximab , Drug Hypersensitivity/diagnosis , Humans , Immunoglobulin E/blood , Infusions, Intravenous/adverse effects , Odds Ratio , Retrospective Studies , Risk Factors
5.
Clin Cancer Res ; 19(6): 1587-95, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23340299

ABSTRACT

PURPOSE: Predictive biomarkers offer the potential to improve the benefit:risk ratio of a therapeutic agent. Ixabepilone achieves comparable pathologic complete response (pCR) rates to other active drugs in the neoadjuvant setting. This phase II trial was designed to investigate potential biomarkers that differentiate response to this agent. EXPERIMENTAL DESIGN: Women with untreated, histologically confirmed primary invasive breast adenocarcinoma received neoadjuvant doxorubicin/cyclophosphamide, followed by 1:1 randomization to ixabepilone (n = 148) or paclitaxel (n = 147). Rates of pCR were compared between treatment arms based on predefined biomarker sets: TUBB3, TACC3, and CAPG gene expression, a 20- and 26-gene expression model, MDR1 protein expression, and other potential markers of sensitivity. ßIII-tubulin protein expression is reported separately but is referred to here for completeness. All patients underwent a core needle biopsy of the primary cancer for molecular marker analysis before chemotherapy. Gene expression profiling data were used for molecular subtyping. RESULTS: There was no significant difference in the rate of pCR in both treatment arms in ßIII-tubulin-positive patients. Higher pCR rates were observed among ßIII-tubulin-positive patients than in ßIII-tubulin-negative patients. Furthermore, no correlation was evident between TUBB3, TACC3, and CAPG gene expression, MDR1 protein expression, multi-gene expression models, and the efficacy of ixabepilone or paclitaxel, even within the estrogen receptor-negative subset. CONCLUSION: These results indicate that ßIII-tubulin protein and mRNA expression, MDR1 protein expression, TACC3 and CAPG gene expression, and multigene expression models (20- and 26-gene) are not predictive markers for differentiating treatment benefit between ixabepilone and paclitaxel in early-stage breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Neoadjuvant Therapy , Neoplasm Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Biomarkers, Tumor , Breast Neoplasms/pathology , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Epothilones/administration & dosage , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Paclitaxel/administration & dosage , Prognosis , Tubulin/genetics
6.
J Clin Oncol ; 31(16): 1990-6, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23589560

ABSTRACT

PURPOSE: Retrospective studies have reported that tumor expression of the beta-3 tubulin (ß3T) isoform is an unfavorable prognostic factor in non-small-cell lung cancer (NSCLC) treated with tubulin-inhibiting chemotherapy. Ixabepilone is a tubulin-inhibiting agent with low susceptibility to multiple resistance mechanisms including ß3T isoform expression in several tumor models. This randomized phase II study evaluated ixabepilone-based chemotherapy in stage IIIb/IV NSCLC, compared with paclitaxel-based chemotherapy. Tumor specimens were prospectively evaluated for ß3T expression. PATIENTS AND METHODS: Patients were stratified by ß3T status (positive v negative) and randomly assigned at a ratio of 1:1 to receive ixabepilone (32 mg/m(2)) and carboplatin (area under concentration-time curve [AUC], 6) or paclitaxel (200 mg/m(2)) and carboplatin (AUC, 6) for up to six cycles. The primary end point was progression-free survival (PFS) in the ß3T-positive subgroup. RESULTS: Ninety-five patients (ß3T positive, 52; ß3T negative, 43) received ixabepilone plus carboplatin; 96 patients (ß3T positive, 49; ß3T negative, 47) received paclitaxel plus carboplatin. No significant differences in median PFS were observed between arms for either subgroup (ß3T positive, 4.3 months in both arms; ß3T negative, 5.8 v 5.3 months). Ixabepilone did not significantly improve overall survival (OS) for the ß3T-positive subset or the overall population. Adverse events were similar between the two arms and comparable with those in previous studies. CONCLUSION: There was no predictive value of ß3T in differentiating clinical activity of ixabepilone- or paclitaxel-containing regimens. Ixabepilone did not improve PFS or OS in patients with ß3T-positive tumors. ß3T-positive patients had worse PFS relative to ß3T-negative patients, regardless of treatment; hence, ß3T expression seems to be a negative prognostic factor, but not a predictive factor, in advanced NSCLC treated with either ixabepilone or paclitaxel platinum-based doublets.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Tubulin/analysis , Adult , Aged , Area Under Curve , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/secondary , Disease-Free Survival , Drug Administration Schedule , Epothilones/administration & dosage , Female , Humans , Lung Neoplasms/chemistry , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Paclitaxel/administration & dosage , Predictive Value of Tests , Prospective Studies , Treatment Outcome , Tubulin Modulators/administration & dosage
7.
Arch Pathol Lab Med ; 137(6): 820-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23030695

ABSTRACT

CONTEXT: The therascreen KRAS RGQ polymerase chain reaction kit is being developed as a companion diagnostic to aid clinicians, through detection of KRAS mutations, in the identification of patients with metastatic colorectal cancer (mCRC) who are more likely to benefit from cetuximab. OBJECTIVES: To assess whether KRAS mutation status, determined by using the therascreen KRAS kit, is a predictive marker of cetuximab efficacy. DESIGN: Tissue samples were obtained from patients with mCRC treated on the National Cancer Institute of Canada Clinical Trials Group (NCIC CTG) CO.17 phase 3 study of cetuximab plus best supportive care (BSC) versus BSC alone. Tumor DNA samples were assessed for the presence of KRAS mutations by using the therascreen KRAS kit. Efficacy and safety were assessed to determine whether mutation status was predictive of outcomes. Results.-Evaluable samples were available from 453 patients (79.2%) enrolled in the NCIC CTG CO.17 trial. The KRAS wild-type subset represented 54.1% (245 of 453) of the evaluated population. Median overall survival of patients with KRAS wild-type tumors was 8.6 months among those who received cetuximab plus BSC and 5.0 months among patients who received BSC alone (hazard ratio [HR], 0.63; P = .002). Among patients with KRAS mutant mCRC, no meaningful difference in overall survival was observed between arms (HR, 0.91; P = .55). These results are consistent with a previous report that analyzed patient tumor samples by using bidirectional sequencing. CONCLUSIONS: These data support the utility of the therascreen KRAS kit as a means of selecting patients who may benefit from cetuximab therapy.


Subject(s)
Adenocarcinoma/secondary , Codon, Nonsense , Codon/genetics , Colorectal Neoplasms/pathology , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Cetuximab , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Mutation , Proto-Oncogene Proteins p21(ras) , Survival Rate
8.
Gastrointest Cancer Res ; 5(5): 155-60, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23112883

ABSTRACT

BACKGROUND: The aim of this phase 2 study was to evaluate the safety and efficacy of ixabepilone plus cetuximab in patients with advanced pancreatic cancer. METHODS: Eligible patients had advanced pancreatic adenocarcinoma that was metastatic or not amenable to resection, a Karnofsky performance status ≥70%, and no prior therapy for advanced disease. Patients received ixabepilone 32 mg/m(2) (3-hour IV infusion) every 3 weeks and cetuximab 250 mg/m(2) (1-hour IV infusion) weekly. The primary efficacy end point was the 6-month survival rate. Secondary end points included tumor response rate, overall survival, progression-free survival, and tolerability. RESULTS: A total of 54 patients were enrolled on this study. The 6-month survival rate was 57% (31/54: 95% CI: 43-71%) with a median overall survival of 7.6 months (95% CI: 5.5-12.2 months). Patients who developed acneiform rash (n = 36) had a median survival of 8.8 months, compared with 2.6 months for those without rash (n = 18). Of 31 patients with measurable disease (defined as response-evaluable), 4 had confirmed partial responses and an additional 24 had stable disease. The combination was generally well-tolerated with the most common grade 3/4 hematological toxicities being leucopenia (39%) and neutropenia (33%). The most common grade 3/4 nonhematologic toxicity was fatigue (17%). CONCLUSIONS: The combination of ixabepilone and cetuximab was active and had acceptable toxicity. The efficacy results are similar to single-agent ixabepilone and gemcitabine-based combination therapies in patients with advanced pancreatic cancer. Exploratory analyses suggest a trend toward improved survival for patients who experienced rash.

9.
Am J Clin Oncol ; 26(4): S58-61, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12902857

ABSTRACT

Prostaglandins have long been known to impact the radiosensitivity of cells and tissues, and many studies have centered on exploiting nonspecific prostaglandin inhibitors such as NSAIDs for therapeutic gain. These studies have ultimately been unsuccessful due to the lack of targeted specificity against the tumor. The discovery of the inducible cyclooxygenase enzyme (COX-2) and development of some highly selective inhibitors (which spare the constitutive COX-1 activity) has renewed excitement for modulating tumor prostaglandins as a method of specific radiosensitization of tumors, while sparing normal tissues. This review discusses these new data and generates a rationale for use of COX-2 inhibitors as radiosensitizing agents in cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Isoenzymes/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Humans , Membrane Proteins , Prostaglandin-Endoperoxide Synthases , Prostaglandins/pharmacology , Prostaglandins/therapeutic use , Radiation-Sensitizing Agents/pharmacology
10.
Proc Natl Acad Sci U S A ; 101(2): 591-6, 2004 Jan 13.
Article in English | MEDLINE | ID: mdl-14688410

ABSTRACT

Overexpression of human cyclooxygenase 2 (COX-2) in the mammary glands of transgenic mice induces tissue-specific tumorigenic transformation. However, the molecular mechanisms involved are not yet defined. Here we show that COX-2 expressed in the epithelial cell compartment regulates angiogenesis in the stromal tissues of the mammary gland. Microvessel density increased before visible tumor growth and exponentially during tumor progression. Inhibition of prostanoid synthesis with indomethacin strongly decreased microvessel density and inhibited tumor progression. Up-regulation of angiogenic regulatory genes in COX-2 transgenic mammary tissue was also potently inhibited by indomethacin treatment, suggesting that prostanoids released from COX-2-expressing mammary epithelial cells induce angiogenesis. G protein-coupled receptors for the major product, prostaglandin E(2) (PGE(2)) EP(1-4), are expressed during mammary gland development, and EP(1,2,4) receptors were up-regulated in tumor tissue. PGE(2) stimulated the expression angiogenic regulatory genes in mammary tumor cells isolated from COX-2 transgenic mice. Such cells are tumorigenic in nude mice; however, treatment with Celecoxib, a COX-2-specific inhibitor, reduced tumor growth and microvessel density. These results define COX-2-derived PGE(2) as a potent inducer of angiogenic switch during mammary cancer progression.


Subject(s)
Dinoprostone/physiology , Isoenzymes/physiology , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/physiopathology , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Chromatography, Liquid , Cyclooxygenase 2 , Disease Progression , Mammary Neoplasms, Experimental/blood supply , Mice , Mice, Nude , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL