Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Front Reprod Health ; 3: 714829, 2021.
Article in English | MEDLINE | ID: mdl-36303974

ABSTRACT

The development of therapies targeted to improve the health of women has utilized direct vaginal delivery as a more effective and less toxic method of protection from HIV and other pathogens. Vaginal applicants and delivery devices that provide sustained effects have been met with increasing acceptability, but the efficacy and toxicity outcomes have not been successfully predicted by preclinical in vitro studies and animal modeling. We have explored the utilization of sheep as a model for testing the safety of vaginal applicants and devices based on spatial and structural similarities to the human vagina. As recently noted by the FDA, an additional safety measure is an impact on the vaginal microbiome (VMB) that is known to contribute to vaginal health and influence pathogen susceptibility and drug metabolism. To advance the utility of the sheep vaginal model, we completed a thorough molecular characterization of the ovine VMB utilizing both next-generation sequencing (NGS) and PCR methods. The process also created a custom PCR array to quantify ovine VMB community profiles in an affordable, higher throughput fashion. The results from vaginal swabs (>475 samples) collected from non-pregnant crossbred Dorset and Merino ewes treated with selected vaginal applicants or collected as sham samples established 16 VMB community types (VMB CTs). To associate VMB CTs with eubiosis or dysbiosis, we also completed custom ELISAs for six cytokines identifying IL1B, IL8, TNFa, and CXCL10 as useful markers to support the characterization of ovine vaginal inflammation. The results indicated that Pasteurella, Actinobacillus, Pseudomonas, Bacteroides, Leptotrichia, and E. coli were common markers of eubiosis (low inflammatory marker expression), and that Haemophilus, Ureaplasma, and Corynebacterium were associated with dysbiosis (high cytokine levels). Utilizing the optimized workflow, we also confirmed the utility of three commonly used vaginal applicants for impact on the VMB and inflammatory state, producing a dataset that supports the recommendation for the use of sheep for testing of vaginal applicants and devices as part of preclinical pipelines.

2.
Front Reprod Health ; 3: 714798, 2021.
Article in English | MEDLINE | ID: mdl-36304006

ABSTRACT

Background: Development of safe, effective products to prevent the sexual transmission of HIV remains a priority. Prior to clinical testing, the products must undergo strict safety evaluations to avoid mucosal drug toxicity, inflammation, and vaginal microbiome (VMB) shifts. Based on the Food and Drug Administration (FDA) guidance, we designed a study to measure the inflammatory markers and VMB changes after intravaginal treatment with products that have been associated with toxicity, with the objective to develop a Gram stain slide scoring system, similar to Nugent scoring, correlated with the proinflammatory cytokines in sheep. Methods: Non-pregnant Dorset ewes (n = 34) were randomized to receive 5 ml intravaginal 4% nonoxynol-9 (N9) contraceptive gel, positive control (0.2% benzalkonium chloride), placebo control [hydroxethyl cellulose (HEC)], or no application daily for 10 days, with 11-day post-treatment follow-up. The vaginal swabs were collected for the cytokines, VMB, and Gram-stained slides. An enzyme-linked immunosorbent assay (ELISA) analysis of cytokines interleukin (IL)-1ß, IL-8, CXCL10, and tumor necrosis factor-α (TNF-α) was used to determine inflammatory state of the sample. Vaginal microbiome community types (CT) were utilized to create five equivalent slide subsets for iterative development of a Gram-stained slide scoring system with comparisons with inflammatory state based on the cytokine levels. Results: Digital images of the Gram-stained slides were scored based on Gram staining and morphology of bacteria, presence of sheep epithelial cells, and immune cells. The scoring system was modified in an iterative fashion with weighting based on cytokine categorization of inflamed samples, with three of four cytokine values above the mean indicating that the sample was inflamed. The parameters in the final version of the scoring system included mature epithelial cells, Gram-negative rods, and Gram-positive diplococci indicating normal and immune cells indicating inflammation. The area under the receiver operator characteristic curve (ROC AUC) was 0.725 (ROC AUCs range between 0.5 and 1.0) with a greater area indicating higher diagnostic ability of a test with a binary outcome: inflamed or normal. Conclusion: The scoring system, derived from the advanced VMB and cytokine analyses, provides a validated, practical method for quantification of Gram-stained slides that can be performed in most laboratories, increasing the potential for standardization. The training plan can assist laboratories to determine the safety of intravaginal products in their sheep studies or the methodological approach can be applied to other animal models where such data are also needed.

3.
Sci Rep ; 7: 41098, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28145472

ABSTRACT

The efficacy of HIV pre-exposure prophylaxis (PrEP) relies on adherence and may also depend on the route of HIV acquisition. Clinical studies of systemic tenofovir disoproxil fumarate (TDF) PrEP revealed reduced efficacy in women compared to men with similar degrees of adherence. To select the most effective PrEP strategies, preclinical studies are critically needed to establish correlations between drug concentrations (pharmacokinetics [PK]) and protective efficacy (pharmacodynamics [PD]). We utilized an in vivo preclinical model to perform a PK-PD analysis of systemic TDF PrEP for vaginal HIV acquisition. TDF PrEP prevented vaginal HIV acquisition in a dose-dependent manner. PK-PD modeling of tenofovir (TFV) in plasma, female reproductive tract tissue, cervicovaginal lavage fluid and its intracellular metabolite (TFV diphosphate) revealed that TDF PrEP efficacy was best described by plasma TFV levels. When administered at 50 mg/kg, TDF achieved plasma TFV concentrations (370 ng/ml) that closely mimicked those observed in humans and demonstrated the same risk reduction (70%) previously attained in women with high adherence. This PK-PD model mimics the human condition and can be applied to other PrEP approaches and routes of HIV acquisition, accelerating clinical implementation of the most efficacious PrEP strategies.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/prevention & control , Pre-Exposure Prophylaxis , Tenofovir/therapeutic use , Animals , Anti-Retroviral Agents/blood , Anti-Retroviral Agents/pharmacokinetics , Dose-Response Relationship, Drug , Emtricitabine/blood , Emtricitabine/pharmacokinetics , Emtricitabine/therapeutic use , Female , HIV Infections/transmission , HIV Infections/virology , HIV-1/genetics , HIV-1/isolation & purification , HIV-1/pathogenicity , Half-Life , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Proportional Hazards Models , RNA, Viral/blood , Tenofovir/blood , Tenofovir/pharmacokinetics
4.
AIDS Res Hum Retroviruses ; 33(4): 395-403, 2017 04.
Article in English | MEDLINE | ID: mdl-27841671

ABSTRACT

The ex vivo challenge assay is being increasingly used as an efficacy endpoint during early human clinical trials of HIV prevention treatments. There is no standard methodology for the ex vivo challenge assay, although the use of different data collection methods and analytical parameters may impact results and reduce the comparability of findings between trials. In this analysis, we describe the impact of data imputation methods, kit type, testing schedule and tissue type on variability, statistical power, and ex vivo HIV growth kinetics. Data were p24 antigen (pg/ml) measurements collected from clinical trials of candidate microbicides where rectal (n = 502), cervical (n = 88), and vaginal (n = 110) tissues were challenged with HIV-1BaL ex vivo. Imputation of missing data using a nonlinear mixed effect model was found to provide an improved fit compared to imputation using half the limit of detection. The rectal virus growth period was found to be earlier and of a relatively shorter duration than the growth period for cervical and vaginal tissue types. On average, only four rectal tissue challenge assays in each treatment and control group would be needed to find a one log difference in p24 to be significant (alpha = 0.05), but a larger sample size was predicted to be needed for either cervical (n = 21) or vaginal (n = 10) tissue comparisons. Overall, the results indicated that improvements could be made in the design and analysis of the ex vivo challenge assay to provide a more standardized and powerful assay to compare efficacy of microbicide products.


Subject(s)
Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Data Collection/methods , HIV Infections/prevention & control , HIV-1/drug effects , HIV-1/growth & development , Specimen Handling/methods , Cervix Uteri/virology , Chemoprevention/methods , Disease Transmission, Infectious/prevention & control , Female , HIV Core Protein p24/analysis , Humans , Pre-Exposure Prophylaxis/methods , Rectum/virology , Reproducibility of Results , Retrospective Studies , Treatment Outcome , Vagina/virology
5.
PLoS One ; 9(3): e93108, 2014.
Article in English | MEDLINE | ID: mdl-24667334

ABSTRACT

AIDS is a global pandemic that has seen the development of novel and effective treatments to improve the quality of life of those infected and reduction of spread of the disease. Palmitic Acid (PA), which we identified and isolated from Sargassum fusiforme, is a naturally occurring fatty acid that specifically inhibits HIV entry by binding to a novel pocket on the CD4 receptor. We also identified a structural analogue, 2-bromopalmitate (2-BP), as a more effective HIV entry inhibitor with a 20-fold increase in efficacy. We have used the structure-activity relationship (SAR) of 2-BP as a platform to identify new small chemical molecules that fit into the various identified active sites in an effort to identify more potent CD4 entry inhibitors. To validate further drug development, we tested the PA and 2-BP scaffold molecules for genotoxic potential. The FDA and International Conference on Harmonisation (ICH) recommends using a standardized 3-test battery for testing compound genotoxicity consisting of the bacterial reverse mutation assay, mouse lymphoma assay, and rat micronucleus assay. PA and 2-BP and their metabolites tested negative in all three genotoxicty tests. 2-BP is the first derivative of PA to undergo pre-clinical screening, which will enable us to now test multiple simultaneous small chemical structures based on activity in scaffold modeling across the dimension of pre-clinical testing to enable transition to human testing.


Subject(s)
Biological Products/chemistry , Biological Products/toxicity , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/toxicity , HIV/drug effects , HIV/physiology , Virus Internalization/drug effects , Animals , Biological Products/pharmacology , Drug Discovery , Female , HIV Fusion Inhibitors/pharmacology , Lymphoma/pathology , Male , Mice , Micronucleus Tests , Palmitates/chemistry , Palmitates/pharmacology , Palmitates/toxicity , Palmitic Acid/chemistry , Palmitic Acid/pharmacology , Palmitic Acid/toxicity , Rats , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Structure-Activity Relationship
6.
Bioorg Med Chem ; 17(20): 7344-52, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19762246

ABSTRACT

A novel series of 17-modified and 2,17-modified analogs of 2-methoxyestradiol (2ME2) were synthesized and characterized. These analogs were designed to retain or potentiate the biological activities of 2ME2 and have diminished metabolic liability. The analogs were evaluated for antiproliferative activity against MDA-MB-231 breast tumor cells, antiangiogenic activity in HUVEC, and estrogenic activity on MCF-7 cell proliferation. Several analogs were evaluated for metabolic stability in human liver microsomes and in vivo in a rat cassette dosing model. This study lead to several 17-modified analogs of 2ME2 that have similar or improved antiproliferative and antiangiogenic activity, lack estrogenic properties and have improved metabolic stability compared to 2ME2.


Subject(s)
Cell Proliferation/drug effects , Estradiol/analogs & derivatives , Estrone/chemical synthesis , Estrone/pharmacology , 2-Methoxyestradiol , Animals , Cell Line , Estradiol/pharmacology , Estrone/chemistry , Humans , Rats , Structure-Activity Relationship
7.
Mol Cancer Ther ; 7(6): 1472-82, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18566218

ABSTRACT

Clinical studies using the microtubule-targeting agent 2-methoxyestradiol (2ME2; Panzem) in cancer patients show that treatment is associated with clinical benefit, including prolonged stable disease, complete and partial responses, and an excellent safety profile. Studies have shown that 2ME2 is metabolized by conjugation at positions 3 and 17 and oxidation at position 17. To define structure-activity relationships for these positions of 2ME2 and to generate metabolically stable analogues with improved anti-tubulin properties, a series of analogues was generated and three lead analogues were selected, ENMD-1198, ENMD-1200, and ENMD-1237. These molecules showed improved metabolic stability with >65% remaining after 2-h incubation with hepatocytes. Pharmacokinetic studies showed that oral administration of the compounds resulted in increased plasma levels compared with 2ME2. All three analogues bind the colchicine binding site of tubulin, induce G(2)-M cell cycle arrest and apoptosis, and reduce hypoxia-inducible factor-1alpha levels. ENMD-1198 and ENMD-1200 showed improved in vitro antiproliferative activities. Significant reductions in tumor volumes compared with vehicle-treated mice were observed in an orthotopic breast carcinoma (MDA-MB-231) xenograft model following daily oral treatment with all compounds (ANOVA, P < 0.05). Significantly improved median survival time was observed with ENMD-1198 and ENMD-1237 (200 mg/kg/d) in a Lewis lung carcinoma metastatic model (P < 0.05). In both tumor models, the high-dose group of ENMD-1198 showed antitumor activity equivalent to that of cyclophosphamide. ENMD-1198 was selected as the lead molecule in this analogue series and is currently in a phase I clinical trial in patients with refractory solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Estrenes/pharmacology , Microtubules/drug effects , Tubulin Modulators/pharmacology , 2-Methoxyestradiol , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Binding, Competitive/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/pharmacology , Drug Screening Assays, Antitumor , Estradiol/analogs & derivatives , Estradiol/chemistry , Estrenes/administration & dosage , Estrenes/chemistry , Estrenes/pharmacokinetics , G2 Phase/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred C57BL , Mitosis/drug effects , Rats , Survival Analysis , Tubulin/metabolism , Tubulin Modulators/administration & dosage , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacokinetics
8.
Clin Cancer Res ; 10(24): 8665-73, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15623651

ABSTRACT

PURPOSE: Head and neck squamous cell carcinomas have been reported to overexpress hypoxia-inducible factor (HIF)-1alpha, a transcription factor that promotes expression of angiogenesis factors and resistance to programmed and therapy-induced cell death. 2-Methoxyestradiol (2ME2) is a natural compound with HIF-1alpha inhibitory activity that is currently being evaluated in phase 1 and 2 clinical trials for advanced solid tumors and multiple myeloma. To our knowledge, this is the first study to evaluate the effects of 2ME2 in head and neck squamous cell carcinoma. EXPERIMENTAL DESIGN: In the present study, we investigated the effects of 2ME2 alone and in combination with paclitaxel, an active agent in recurrent or advanced head and neck squamous cell carcinoma. RESULTS: 2ME2 exhibited antiproliferative and cytotoxic effects in a panel of five head and neck squamous cell carcinoma cell lines in the 0.5 to 10 micromol/L range, including induction of G2-M blockade, caspase-3/7 activation, and apoptosis at 48 hours. 2ME2 resulted in decreased nuclear HIF-1alpha-binding activity and affected the expression of downstream genes, such as bid, a proapoptotic bcl-2 family member, and vascular endothelial growth factor, a proangiogenic cytokine. The up-regulation of Bid (57.5% at 12 hours, P < 0.0006) and inhibition of vascular endothelial growth factor secretion (57.7% at 24 hours, P < 0.015; and 50.3% at 48 hours, P < 0.0006) could be partially attributed to the effects on HIF-1alpha, because HIF-1alpha small interfering RNAs produced similar effects. Finally, in vivo, in a xenograft model of head and neck squamous cell carcinoma using UM-SCC-11A cells, 2ME2 exhibited antitumor and antiangiogenic activity, as measured by CD31 immunostaining. CONCLUSIONS: These results provide support for the use of 2ME2 in combination with paclitaxel for the treatment of recurrent or advanced head and neck squamous cell carcinoma.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Squamous Cell/prevention & control , Cell Hypoxia , Estradiol/analogs & derivatives , Head and Neck Neoplasms/prevention & control , Paclitaxel/therapeutic use , Transcription Factors/metabolism , 2-Methoxyestradiol , Animals , BH3 Interacting Domain Death Agonist Protein , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carrier Proteins/metabolism , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Division/drug effects , Drug Therapy, Combination , Enzyme Activation/drug effects , Estradiol/therapeutic use , Female , G2 Phase/drug effects , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Mice, Inbred BALB C , Mice, SCID , Microcirculation , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Blood ; 103(9): 3374-80, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14739228

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is the major physiologic inhibitor of the extrinsic coagulation pathway. We have previously shown that TFPI is also a potent inhibitor of endothelial proliferation in vitro and of primary and metastatic tumor growth in vivo. Surprisingly, the antitumor activity of TFPI was demonstrated to be independent of its anticoagulant activity, suggesting a possible nonhemostatic mechanism of action for TFPI in these models. This antitumor mechanism may involve the very low density lipoprotein (VLDL) receptor because the in vitro antiproliferative activity of TFPI is mediated through interaction with the VLDL receptor. In the current study, we identify a 23-amino acid fragment of TFPI (TFPIc23) localized to the C-terminus, which mediates binding to the VLDL receptor. The TFPIc23 peptide inhibits endothelial cell proliferation through an apoptotic mechanism and blocks vessel outgrowth in the in vitro assays, and this activity is mediated through interaction with the VLDL receptor. In vivo, this peptide potently inhibits angiogenesis in Matrigel and chick chorioallantoic membrane models and also inhibits metastatic tumor growth. Our data demonstrate that this VLDL receptor-binding fragment of the TFPI molecule has apoptotic, antiangiogenic, and antitumor activity and suggests a possible mechanism whereby TFPI can regulate angiogenesis and tumor growth independently of its anticoagulant activity.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Lipoproteins/pharmacology , Peptide Fragments/pharmacology , Receptors, LDL/metabolism , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Binding Sites , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Cell Division/drug effects , Chick Embryo , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Lipoproteins/chemistry , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/prevention & control , Neovascularization, Physiologic/drug effects , Peptide Fragments/metabolism , Umbilical Veins/cytology
10.
Cancer Res ; 63(11): 2997-3000, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12782609

ABSTRACT

An association between cancer and thrombosis has been recognized for more than a century. However, the manner by which tumor growth is regulated by coagulation in vivo remains unclear. To assess the role of coagulation on tumor growth, in vivo, we tested coagulation inhibitors specific for either tissue factor (TF)/factor VIIa (fVIIa) complexes or factor Xa (fXa) for antitumor activity. Here, we show that two inhibitors of TF/fVIIa, TF pathway inhibitor (TFPI) and the nematode anticoagulant protein rNAPc2, inhibit both primary and metastatic tumor growth in mice. In addition, we show that rNAPc2 is also a potent inhibitor of angiogenesis. In contrast, rNAP5, a second nematode anticoagulant protein that specifically inhibits fXa, does not exhibit antitumor activity. Because the hemostatic activity of TF/fVIIa is mediated through activation of fXa, these data suggest that proteolytic activity of TF/fVIIa promotes tumor growth and angiogenesis through a novel proangiogenic mechanism and independently of hemostasis.


Subject(s)
Factor VIIa/antagonists & inhibitors , Helminth Proteins/pharmacology , Lipoproteins/pharmacology , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Thromboplastin/antagonists & inhibitors , Animals , Anticoagulants/pharmacology , Cell Division/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/prevention & control
11.
Cancer Cell ; 3(4): 363-75, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12726862

ABSTRACT

Inhibition of angiogenesis is an important new modality for cancer treatment. 2-methoxyestradiol (2ME2) is a novel antitumor and antiangiogenic agent, currently in clinical trials, whose molecular mechanism of action remains unclear. Herein, we report that 2ME2 inhibits tumor growth and angiogenesis at concentrations that efficiently disrupt tumor microtubules (MTs) in vivo. Mechanistically, we found that 2ME2 downregulates hypoxia-inducible factor-1 (HIF) at the posttranscriptional level and inhibits HIF-1-induced transcriptional activation of VEGF expression. Inhibition of HIF-1 occurs downstream of the 2ME2/tubulin interaction, as disruption of interphase MTs is required for HIF-alpha downregulation. These data establish 2ME2 as a small molecule inhibitor of HIF-1 and provide a mechanistic link between the disruption of the MT cytoskeleton and inhibition of angiogenesis.


Subject(s)
DNA-Binding Proteins/drug effects , Estradiol/pharmacology , Microtubules/drug effects , Neovascularization, Pathologic , Nuclear Proteins/drug effects , Transcription Factors , 2-Methoxyestradiol , Animals , Blotting, Northern , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , DNA-Binding Proteins/metabolism , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Estradiol/analogs & derivatives , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/drug effects , Lymphokines/genetics , Lymphokines/metabolism , Mice , Microscopy, Confocal , Models, Animal , Nuclear Proteins/metabolism , RNA, Messenger , Transcription, Genetic , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Cancer Res ; 63(2): 468-75, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12543804

ABSTRACT

2-Methoxyestradiol (2ME2), a natural metabolite of estradiol, is a potent antitumor and antiangiogenic agent. In vitro, 2ME2 inhibits the proliferation of a wide variety of cell lines and primary cultures, and in numerous models in vivo, it has been shown to be an effective inhibitor of tumor growth and angiogenesis. 2ME2 is currently in several Phase I and Phase II clinical trials under the name Panzem. Although various molecular targets have been proposed for this compound, the mechanism by which 2ME2 exerts its effects is still uncertain. This study shows that 2ME2 uses the extrinsic pathway for induction of apoptosis. 2ME2 treatment results in up-regulation of death receptor 5 (DR5) protein expression in vitro and in vivo and renders cells more sensitive to the cytotoxic activities of the DR5 ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). 2ME2-induced apoptosis requires caspase activation and kinetic studies show the sequential activation of caspase-8, caspase-9, and caspase-3. Blockage of death receptor signaling by expression of dominant-negative Fas-associated death domain severely attenuates the ability of 2ME2 to induce apoptosis. Because 2ME2 administration has not manifested dose-limiting toxicity in the clinic, DR5 expression may serve as a surrogate marker for biological response.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Estradiol/pharmacology , Receptors, Tumor Necrosis Factor/physiology , 2-Methoxyestradiol , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/physiology , Caspases/metabolism , Cell Division/drug effects , Drug Synergism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Activation , Estradiol/analogs & derivatives , Fas-Associated Death Domain Protein , Female , Humans , Isoenzymes/metabolism , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...