Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Lancet Oncol ; 19(7): 904-915, 2018 07.
Article in English | MEDLINE | ID: mdl-29804902

ABSTRACT

BACKGROUND: In MONALEESA-2, ribociclib plus letrozole showed improved progression-free survival compared with letrozole alone as first-line treatment for postmenopausal patients with hormone receptor (HR)-positive, HER2-negative, advanced breast cancer. MONALEESA-7 aimed to assess the efficacy and safety of ribociclib plus endocrine therapy in premenopausal women with advanced, HR-positive breast cancer. METHODS: This phase 3, randomised, double-blind, placebo-controlled trial was done at 188 centres in 30 countries. Eligible patients were premenopausal women aged 18-59 years who had histologically or cytologically confirmed HR-positive, HER2-negative, advanced breast cancer; an Eastern Cooperative Oncology Group performance status of 0 or 1; measurable disease as per Response Evaluation Criteria in Solid Tumors version 1.1 criteria, or at least one predominantly lytic bone lesion; and had not received previous treatment with cyclin-dependent kinases 4 and 6 inhibitors. Endocrine therapy and chemotherapy in the adjuvant or neoadjuvant setting was permitted, as was up to one line of chemotherapy for advanced disease. Patients were randomly assigned (1:1) via interactive response technology to receive oral ribociclib (600 mg/day on a 3-weeks-on, 1-week-off schedule) or matching placebo with either oral tamoxifen (20 mg daily) or a non-steroidal aromatase inhibitor (letrozole 2·5 mg or anastrozole 1 mg, both oral, daily), all with goserelin (3·6 mg administered subcutaneously on day 1 of every 28-day cycle). Patients and investigators were masked to treatment assignment. Efficacy analyses were by intention to treat, and safety was assessed in all patients who received at least one dose of any study treatment. The primary endpoint was investigator-assessed progression-free survival. MONALEESA-7 is registered with ClinicalTrials.gov, NCT02278120 and is ongoing, but no longer enrolling patients. FINDINGS: Between Dec 17, 2014, and Aug 1, 2016, 672 patients were randomly assigned: 335 to the ribociclib group and 337 to the placebo group. Per investigator's assessment, median progression-free survival was 23·8 months (95% CI 19·2-not reached) in the ribociclib group compared with 13·0 months (11·0-16·4) in the placebo group (hazard ratio 0·55, 95% CI 0·44-0·69; p<0·0001). Grade 3 or 4 adverse events reported in more than 10% of patients in either group were neutropenia (203 [61%] of 335 patients in the ribociclib group and 12 [4%] of 337 in the placebo group) and leucopenia (48 [14%] and four [1%]). Serious adverse events occurred in 60 (18%) of 335 patients in the ribociclib group and 39 (12%) of 337 in the placebo group, of which 15 (4%) and six (2%), respectively, were attributed to the study regimen. 12 (4%) of 335 patients in the ribociclib group and ten (3%) of 337 in the placebo group discontinued treatment because of adverse events. No treatment-related deaths occurred. 11 deaths occurred (five [1%] in the ribociclib group and six [2%] in the placebo group) during or within 30 days after treatment, most of which were due to progression of the underlying breast cancer (three [1%] and six [2%]). The remaining two deaths in the ribociclib group were due to an intracranial haemorrhage in an anticoagulated patient, and a pre-existing wound haemorrhage in another patient. INTERPRETATION: Ribociclib plus endocrine therapy improved progression-free survival compared with placebo plus endocrine therapy, and had a manageable safety profile in patients with premenopausal, HR-positive, HER2-negative, advanced breast cancer. The combination could represent a new first-line treatment option for these patients. FUNDING: Novartis.


Subject(s)
Aminopyridines/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Purines/administration & dosage , Administration, Oral , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease-Free Survival , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Humans , Internationality , Kaplan-Meier Estimate , Letrozole/administration & dosage , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Premenopause/drug effects , Prognosis , Proportional Hazards Models , Survival Analysis , Tamoxifen/administration & dosage , Treatment Outcome , Young Adult
2.
Regul Toxicol Pharmacol ; 95: 17-28, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29496518

ABSTRACT

Based on 13 chronic studies, styrene exposure causes lung tumors in mice, but no tumor increases in other organs in mice or rats. Extensive research into the mode of action demonstrates the key events and human relevance. Key events are: metabolism of styrene by CYP2F2 in mouse lung club cells to ring-oxidized metabolites; changes in gene expression for metabolism of lipids and lipoproteins, cell cycle and mitotic M-M/G1 phases; cytotoxicity and mitogenesis in club cells; and progression to preneoplastic/neoplastic lesions in lung. Although styrene-7,8-oxide (SO) is a common genotoxic styrene metabolite in in vitro studies, the data clearly demonstrate that SO is not the proximate toxicant and that styrene does not induce a genotoxic mode of action. Based on complete attenuation of styrene short-term and chronic toxicity in CYP2F2 knockout mice and similar attenuation in CYP2F1 (humanized) transgenic mice, limited metabolism of styrene in human lung by CYP2F1, 2 + orders of magnitude lower SO levels in human lung compared to mouse lung, and lack of styrene-related increase in lung cancer in humans, styrene does not present a risk of cancer to humans.


Subject(s)
Carcinogens/toxicity , Lung Neoplasms/chemically induced , Styrene/toxicity , Animals , Carcinogens/pharmacokinetics , Cell Survival/drug effects , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , Humans , Lipid Metabolism/genetics , Lung/drug effects , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice, Knockout , Rats , Risk Assessment , Species Specificity , Styrene/pharmacokinetics
3.
Cancer Chemother Pharmacol ; 78(5): 921-927, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27681579

ABSTRACT

PURPOSE: A capsule formulation of the tyrosine kinase inhibitor dovitinib (TKI258) was recently studied in a phase 3 renal cell carcinoma trial; however, tablets are the planned commercial formulation. Therefore, this randomized 2-way crossover study evaluated the bioequivalence of dovitinib tablet and capsule formulations in pretreated patients with advanced solid tumors, excluding breast cancer. METHODS: In this 2-part study, eligible patients received dovitinib 500 mg once daily on a 5-days-on/2-days-off schedule. During the 2-period bioequivalence phase, patients received their initial formulation (capsule or tablet) for 3 weeks before being switched to the alternative formulation in the second period. Patients could continue to receive dovitinib capsules on the same dosing schedule during the post-bioequivalence phase. RESULTS: A total of 173 patients were enrolled into the bioequivalence phase of the study (capsule â†’ tablet, n = 88; tablet â†’ capsule, n = 85), and 69 patients had evaluable pharmacokinetics (PK) for both periods. PK analyses showed similar exposure and PK profiles for the dovitinib capsule and tablet formulations and supported bioequivalence [geometric mean ratios: AUClast, 0.95 (90 % CI 0.88-1.01); C max, 0.98 (90 % CI 0.91-1.05)]. The most common adverse events, suspected to be study drug related, included diarrhea (60 %), nausea (53 %), fatigue (45 %), and vomiting (43 %). Of 168 patients evaluable for response, 1 achieved a partial response, and stable disease was observed in 32 % of patients. CONCLUSIONS: Dovitinib capsules and tablets were bioequivalent, with a safety profile similar to that observed in other dovitinib studies of patients with heavily pretreated advanced solid tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Benzimidazoles/administration & dosage , Benzimidazoles/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinolones/administration & dosage , Quinolones/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Benzimidazoles/adverse effects , Capsules , Cross-Over Studies , Demography , Drug Compounding , Female , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quinolones/adverse effects , Tablets , Therapeutic Equivalency , Treatment Outcome , Young Adult
4.
Cryobiology ; 72(3): 225-31, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27074589

ABSTRACT

This study evaluates the effect of undissolved air on isochoric freezing of aqueous solutions. Isochoric freezing is concerned with freezing in a constant volume thermodynamic system. A possible advantage of the process is that it substantially reduces the percentage of ice in the system at every subzero temperature, relative to atmospheric freezing. At the pressures generated by isochoric freezing, or high pressure isobaric freezing, air cannot be considered an incompressible substance and the presence of undissolved air substantially increases the amount of ice that forms at any subfreezing temperature. This effect is measurable at air volumes as low as 1%. Therefore eliminating the undissolved air, or any separate gaseous phase, from the system is essential for retaining the properties of isochoric freezing.


Subject(s)
Air , Cryopreservation , Freezing , Ice , Pressure , Temperature , Thermodynamics
5.
Toxicology ; 294(2-3): 104-8, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22386858

ABSTRACT

Styrene causes toxicity in both the lung and the liver. The study of the relationship of this toxicity to the metabolism of styrene has been aided by the use of knockout mice for both bioactivation and detoxification pathways. It has been hypothesized that CYP2E1 is primarily responsible for styrene bioactivation in mouse liver and CYP2F2 in mouse lung. Two knockout strains were used in the current studies. Mice deficient in hepatic cytochrome P450 reductase had much less hepatic metabolism of styrene to styrene oxide. Styrene (600 mg/kg, i.p.) caused significant hepatotoxicity, as determined by serum sorbitol dehydrogenase and glutathione levels, in the wild-type but not in the knockout mice. It caused lung toxicity, as determined by protein levels, cell number, and lactate dehydrogenase activity in the bronchioalveolar lavage fluid of wild-type mice, but this effect was less in the knockout mice. In CYP2F2 knockout mice there was only a small decrease in the hepatic metabolism of styrene but a very large decrease in pulmonary metabolism. As expected the CYP2F2 knockout and wild-type mice were equally susceptible to styrene-induced hepatotoxicity, but the knockout mice were less susceptible to styrene-induced pneumotoxicity. Although the results are inconsistent with the simple hypothesis that styrene pneumotoxicity is due to the bioactivation of styrene to styrene oxide by CYYP2F2, they demonstrate the importance of both liver and lung in the metabolism of styrene, but additional pharmacokinetic studies are needed to help clarify the relationship between target organ metabolism and susceptibility.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Epoxy Compounds/toxicity , Liver/drug effects , NADPH-Ferrihemoprotein Reductase/genetics , Styrene/toxicity , Animals , Bronchoalveolar Lavage Fluid , Epoxy Compounds/metabolism , Glutathione/metabolism , L-Iditol 2-Dehydrogenase/blood , Liver/enzymology , Liver/metabolism , Lung/drug effects , Lung/enzymology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Styrene/metabolism
6.
Toxicol Sci ; 127(2): 331-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22461448

ABSTRACT

Toxicology and careers in toxicology, as well as many other scientific disciplines, are undergoing rapid and dramatic changes as new discoveries, technologies, and hazards advance at a blinding rate. There are new and ever increasing demands on toxicologists to keep pace with expanding global economies, highly fluid policy debates, and increasingly complex global threats to public health. These demands must be met with new paradigms for multidisciplinary, technologically complex, and collaborative approaches that require advanced and continuing education in toxicology and associated disciplines. This requires paradigm shifts in educational programs that support recruitment, development, and training of the modern toxicologist, as well as continued education and retraining of the midcareer professional to keep pace and sustain careers in industry, government, and academia. The Society of Toxicology convened the Toxicology Educational Summit to discuss the state of toxicology education and to strategically address educational needs and the sustained advancement of toxicology as a profession. The Summit focused on core issues of: building for the future of toxicology through educational programs; defining education and training needs; developing the "Total Toxicologist"; continued training and retraining toxicologists to sustain their careers; and, finally, supporting toxicology education and professional development. This report summarizes the outcomes of the Summit, presents examples of successful programs that advance toxicology education, and concludes with strategies that will insure the future of toxicology through advanced educational initiatives.


Subject(s)
Education, Professional/trends , Toxicology/education , Toxicology/trends , Cooperative Behavior , Curriculum/trends , Fellowships and Scholarships/trends , Forecasting , Humans , Interinstitutional Relations , Needs Assessment/trends , Professional Competence , Research Support as Topic/trends
7.
Drug Chem Toxicol ; 34(4): 440-4, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21749217

ABSTRACT

Styrene is known to be hepatotoxic and pneumotoxic in rodents, and these adverse effects are related to its metabolism. Mice deficient in the enzymes responsible for both the activation and detoxification of styrene are useful in examining this relationship more closely. In the current study, mice deficient in glutathione S-transferase P1P2(-/-) (GST(-/-)) were compared with wild-type mice. Similar changes in serum sorbitol dehydrogenase, as an indicator of hepatotoxicity, and bronchioalveolar levels of protein, cells, and lactate dehydrogenase, as indicators of pneumotoxicity, were observed after styrene administration. Glutathione depletion followed a similar pattern. The administration of the toxic metabolite, styrene oxide, which is a direct substrate for glutathione metabolism, and 4-vinylphenol, which is a minor metabolite but is more potent than either styrene oxide, yielded results similar to those of styrene. The results indicate that either other isoforms of glutathione S-transferase are more important than the P1P2 form in styrene detoxification or that this pathway contributes in only a minor way to styrene detoxification, compared to other pathways.


Subject(s)
Epoxy Compounds/toxicity , Glutathione Transferase/deficiency , Liver/drug effects , Lung/drug effects , Phenols/toxicity , Styrene/toxicity , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Data Interpretation, Statistical , Glutathione/metabolism , Glutathione Transferase/genetics , Inactivation, Metabolic , Liver/enzymology , Liver/metabolism , Liver/pathology , Lung/enzymology , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Oxidative Stress/drug effects , Styrene/metabolism
8.
J Toxicol Environ Health A ; 74(6): 347-50, 2011.
Article in English | MEDLINE | ID: mdl-21271435

ABSTRACT

Styrene is hepatotoxic and pneumotoxic in mice. Styrene oxide, the active metabolite, is detoxified via hydrolysis by microsomal epoxide hydrolase (mEH). Racemic styrene oxide was previously found to be more lethal and produced increased toxicity in mEH-/- mice compared to wild-type mice. The hepatotoxicity and pneumotoxicity of the R- and S-styrene oxide (SO) enantiomers were compared in wild-type and mEH-deficient mice (mEH-/-). Twenty-four hours following administration of 150 mg/kg ip, neither enantiomer produced hepatotoxicity, but S-SO was more pneumotoxic. However, in mEH-/- mice R-SO produced greater decreases in hepatic glutathione levels 3 h after administration. The basis for the unusual greater toxicity of S-SO, rather than the generally more toxic R-SO, in mEH-/- mice may be related to differences in detoxification by EH.


Subject(s)
Disease Models, Animal , Epoxide Hydrolases/deficiency , Epoxy Compounds/metabolism , Epoxy Compounds/toxicity , Liver/drug effects , Lung/drug effects , Microsomes, Liver/drug effects , Animals , Epoxide Hydrolases/metabolism , Epoxy Compounds/administration & dosage , Glutathione/analysis , Glutathione/metabolism , Inactivation, Metabolic , Liver/enzymology , Liver/metabolism , Lung/enzymology , Lung/metabolism , Mice , Mice, Inbred C57BL , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Stereoisomerism , Time Factors
9.
J Toxicol Environ Health A ; 73(24): 1689-99, 2010.
Article in English | MEDLINE | ID: mdl-21058172

ABSTRACT

Styrene, which is widely used in manufacturing, is both acutely and chronically toxic to mice. Styrene is metabolized by cytochromes P-450 to the toxic metabolite styrene oxide, which is detoxified via hydrolysis with microsomal epoxide hydrolase (mEH) playing a major role. The purpose of these studies was to characterize the importance of this pathway by determining the hepatotoxicity and pneumotoxicity of styrene in wild-type and mEH-deficient (mEH(-/-)) mice. While the mEH(-/-) mice metabolized styrene to styrene oxide at the same rate as the wild-type mice, as expected there was minimal metabolism of styrene oxide to glycol. mEH(-/-) mice were more susceptible to the lethal effects of styrene. Twenty-four hours following the administration of 200 mg/kg ip styrene, mice demonstrated a greater hepatotoxic response due to styrene, as measured by increased serum sorbitol dehydrogenase activity and greater pneumotoxicity as shown by increased protein levels, cell numbers, and lactate dehydrogenase activity in bronchioalveolar lavage fluid. mEH(-/-) mice were also more susceptible to styrene-induced oxidative stress, as indicated by greater decreases in hepatic glutathione levels 3 h after styrene. Styrene oxide at a dose of 150 mg/kg did not produce hepatotoxicity in either wild-type or mEH(-/-) mice. However, styrene oxide produced pneumotoxicity that was similar in the two strains. Thus, mEH plays an important role in the detoxification of styrene but not for exogenously administered styrene oxide.


Subject(s)
Bronchi/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Epoxide Hydrolases/deficiency , Microsomes/drug effects , Styrene/metabolism , Styrene/toxicity , Animals , Bronchi/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemical and Drug Induced Liver Injury/enzymology , Dose-Response Relationship, Drug , Epoxide Hydrolases/metabolism , Epoxy Compounds , Glutathione/metabolism , L-Iditol 2-Dehydrogenase/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Microsomes/metabolism , Styrene/administration & dosage , Time Factors
10.
J Toxicol Environ Health A ; 73(11): 766-72, 2010.
Article in English | MEDLINE | ID: mdl-20391119

ABSTRACT

Styrene is a widely used chemical, but it is known to produce lung and liver damage in mice. This may be related to oxidative stress associated with the decrease in the levels of reduced glutathione (GSH) in the target tissues. The purpose of this study was to investigate the effect of styrene and its primary metabolites R-styrene oxide (R-SO) and S-styrene oxide (S-SO) on GSH levels in the lung lumen, as determined by amounts of GSH in bronchioalveolar lavage fluid (BALF) and in plasma. When non-Swiss albino (NSA) mice were administered styrene (600 mg/kg, ip), there was a significant fall in GSH levels in both BALF and plasma within 3 h. These returned to control levels by 12 h. The active metabolite R-SO (300 mg/kg, ip) also produced significant decreases in GSH in both BALF and plasma, but S-SO was without marked effect. Since GSH is a principal antioxidant in the lung epithelial lining fluid, this fall due to styrene may exert a significant influence on the ability of the lung to buffer oxidative damage.


Subject(s)
Air Pollutants, Occupational/toxicity , Bronchoalveolar Lavage Fluid/chemistry , Epoxy Compounds/toxicity , Glutathione/blood , Lung/drug effects , Oxidative Stress/drug effects , Styrene/toxicity , Animals , Disease Models, Animal , Injections, Intraperitoneal , Lung/metabolism , Male , Mice , Time Factors
11.
Toxicology ; 264(3): 171-8, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19666080

ABSTRACT

In mice, styrene is hepatotoxic, pneumotoxic, and causes lung tumors. One explanation for the mechanism of toxicity is oxidative stress/damage. Previous studies have shown decreased glutathione levels, linked to increased apoptosis, in lung homogenates and isolated Clara cells 3 h following styrene or styrene oxide (SO) administration or in vitro exposure. The objective of the current studies was to determine what effects styrene and its active metabolites, primarily styrene oxide, had on indicators of oxidative stress and attendant apoptosis in order to understand better the mechanism of styrene-induced toxicity. Three hours following in vitro exposure of Clara cells to styrene or SO there were increases in reactive oxygen species (ROS). Following administration of styrene or styrene oxide ip, increases in ROS, superoxide dismutase (SOD), and 8-hydroxydeoxyguanosine (8-OHdG) formation were observed. Since increases in ROS have been linked to increases in apoptosis ratios of bax/bcl-2, mRNA and protein expression were determined 3-240 h following the administration of styrene and R-styrene oxide (RSO). The bax/bcl-2 mRNA ratio increased 12 and 24 h following R-SO and 120 h following styrene administration. However, the bax/bcl-2 protein ratio was not increased until 240 h following R-SO, and 24 and 240 h following styrene administration. However, only a slight increase in caspase 3 was observed. These results indicated that oxidative stress occurred 3h following styrene or styrene oxide as evidenced by increased ROS and SOD. This increased ROS may be responsible for the increased 8-OHdG formation. Our findings of limited apoptosis in Clara cells following acute exposure to styrene or SO are in agreement with others and may reflect the minimal extent to which apoptosis plays a role in acute styrene toxicity. It is clear, however, that oxidative stress and oxidative effects on DNA are increased following exposure to styrene or styrene oxide, and these may play a role in the lung tumorigenesis in mice.


Subject(s)
Apoptosis/drug effects , Carcinogens/toxicity , Epithelial Cells/drug effects , Epoxy Compounds/toxicity , Lung/drug effects , Oxidative Stress/drug effects , Styrene/toxicity , 8-Hydroxy-2'-Deoxyguanosine , Animals , Biomarkers/metabolism , Carcinogens/administration & dosage , Carcinogens/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Cells, Cultured , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epoxy Compounds/administration & dosage , Epoxy Compounds/metabolism , Injections, Intraperitoneal , Lung/metabolism , Lung/pathology , Male , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Styrene/administration & dosage , Styrene/metabolism , Superoxide Dismutase/metabolism , Time Factors , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
12.
Regul Toxicol Pharmacol ; 55(2): 205-18, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19589367

ABSTRACT

It is proposed that metabolism of several structurally-related chemicals by CYP2F isoforms of the cytochromes P450 family results in a cytotoxicity-driven mode of action in organs high in CYP2F; namely, CYP2F2 in nasal and lung tissue in mice and CYP2F4 in nasal tissues in rats. Importantly, the CYP2F1 isozyme expressed in humans appears to have a low capacity to metabolize these compounds. In mice, the resultant cytotoxicity and subsequent regenerative hyperplasia is hypothesized drive an increase in lung tumors that are mostly benign and are not life shortening. Although a complete picture of the mode of action has not been developed in any one model compound, data from the individual compounds can be combined to synthesize and reinforce confidence in the CYP2F toxicity hypothesis. For coumarin, naphthalene, and styrene, inhibition of toxicity with inhibition of CYP2F2 has been demonstrated. Rat CYP2F4 appears to be equally active in metabolizing these chemicals; however, CYP2F4 occurs to a much lower extent in rat Clara cells and levels of metabolites produced are not sufficient to cause lung cytotoxicity. Human lungs contain far fewer of Clara cells than rats or mice, and human lung microsomes failed to, or only marginally, metabolize these compounds. In addition, the human lung differs markedly from the mouse lung in the morphology of its Clara cells, which make humans much less sensitive than mice to toxicity due to reactive metabolites. The absence of a role for CYP2E1-generated metabolites (primarily alkyl oxidation vs. ring-oxidation) in mouse pulmonary effects was demonstrated by the lack of protection from styrene toxicity by CYP2E1 inhibitor, or reduction of toxicity in CYP2E1-knockout mice, and lack of lung toxicity of the primary metabolite of ethylbenzene. The chemicals used as examples of this mode of action generally are negative in standard genotoxicity assays. Apart from increased SCE, no consistent pattern in genotoxicity results was found among these chemicals. Thus, while lung tumors from bronchiolar cell cytotoxicity are theoretically possible in humans, it is unlikely that metabolism by CYP2F1 would produce levels of cytotoxic metabolites in human lungs sufficient to result in lung cytotoxic responses and thus tumors. Therefore, it is unlikely several chemicals that cause mouse lung tumors via CYP2F2 metabolism will cause lung tumors in humans.


Subject(s)
Adenocarcinoma/chemically induced , Adenoma/chemically induced , Bronchioles/drug effects , Carcinogens/toxicity , Cytochrome P-450 Enzyme System/metabolism , Lung Neoplasms/chemically induced , Adenocarcinoma/enzymology , Adenoma/enzymology , Animals , Bronchioles/enzymology , Bronchioles/pathology , Carcinogens/chemistry , Carcinogens/metabolism , Cell Proliferation/drug effects , Coumarins/chemistry , Coumarins/metabolism , Coumarins/toxicity , Cytochrome P-450 Enzyme Inhibitors , Disease Models, Animal , Endpoint Determination , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Inhalation Exposure , Isoenzymes , Lung Neoplasms/enzymology , Mice , Mice, Knockout , Mutagenicity Tests , Naphthalenes/chemistry , Naphthalenes/metabolism , Naphthalenes/toxicity , Rats , Structure-Activity Relationship , Styrene/chemistry , Styrene/metabolism , Styrene/toxicity
13.
Toxicology ; 259(3): 149-52, 2009 May 17.
Article in English | MEDLINE | ID: mdl-19428955

ABSTRACT

Styrene exposure is highest among workers in the reinforced plastics industry with exposure seen for 5 consecutive days during the work week. Styrene is both hepatotoxic and pneumotoxic in mice, in addition to causing lung tumors. Human epidemiological studies are inconclusive as to the carcinogenicity of styrene so it is important to understand the mechanism responsible for styrene tumors in mice. Previous studies showed significant decreases in CC10 protein for 5 days following a single dose of the active metabolite R-styrene oxide (R-SO), yet little change in the bax/bcl-2 protein ratio was seen until 10 days following styrene or R-SO administration. Styrene or R-SO was given to CD-1 mice for 5 consecutive days. Mice were euthanized 24h, 10 days or 30 days following the last dose, and CC10, bax and bcl-2 mRNA and protein levels were determined in isolated Clara cells. CC10 mRNA levels were decreased at 24h for both styrene and R-SO. R-SO decreased CC10 protein levels up to 10 days following the last dose. Increases in the bax/bcl-2 mRNA and protein ratio were seen 24h following R-SO administration. Styrene did not significantly increase the bax/bcl-2 mRNA ratio until 10 days after treatment, with the bax/bcl-2 protein ratio increased at both 10 days and 30 days. It is likely that oxidative stress is involved in the toxicity caused by styrene and that minimal apoptosis may be involved. Chronically decreased CC10 levels may lead to increases in oxidative stress in Clara cells, the main target for styrene toxicity in the lung, and may be an early indicator for lung carcinogenesis in mice.


Subject(s)
Epoxy Compounds/toxicity , Lung/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Styrene/toxicity , Uteroglobin/biosynthesis , bcl-2-Associated X Protein/biosynthesis , Animals , Blotting, Western , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Lung/cytology , Lung/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Male , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Uteroglobin/genetics , bcl-2-Associated X Protein/genetics
14.
J Toxicol Environ Health A ; 72(10): 642-50, 2009.
Article in English | MEDLINE | ID: mdl-19308849

ABSTRACT

Styrene produces lung and liver damage that may be related to oxidative stress. The purpose of this study was to investigate the toxicity of (R)-styrene oxide (R-SO), the more active enantiomeric metabolite of styrene, and the protective properties of the antioxidants glutathione (GSH), N-acetylcysteine (NAC), and 4-methoxy-L-tyrosinyl-gamma-L-glutamyl-L-cysteinyl-glycine (UPF1) against R-SO-induced toxicity in non-Swiss Albino (NSA) mice. UPF1 is a synthetic GSH analog that was shown to have 60 times the ability to scavenge reactive oxygen species (ROS) in comparison to GSH. R-SO toxicity to the lung was measured by elevations in the activity of lactate dehydrogenase (LDH), protein concentration, and number of cells in bronchoalveolar lavage fluid (BALF). Toxicity to the liver was measured by increases in serum sorbitol dehydrogenase (SDH) activity. Antioxidants were not able to decrease the adverse effects of R-SO on lung. However, NAC (200 mg/kg) ip and GSH (600 mg/kg), administered orally prior to R-SO (300 mg/kg) ip, showed significant protection against liver toxicity as measured by SDH activity. Unexpectedly, a synthetic GSH analog, UPF1 (0.8 mg/kg), administered intravenously (iv) prior to R-SO, produced a synergistic effect with regard to liver and lung toxicity. Treatment with UPF1 (0.8 mg/kg) iv every other day for 1 wk for preconditioning prior to R-SO ip did not result in any protection against liver and lung toxicity, but rather enhanced the toxicity when administered prior R-SO. The results of the present study demonstrated protection against R-SO toxicity in liver but not lung by the administration of the antioxidants NAC and GSH.


Subject(s)
Antioxidants/pharmacology , Epoxy Compounds/toxicity , Oxidative Stress/drug effects , Acetylcysteine/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Chemical and Drug Induced Liver Injury/pathology , Epoxy Compounds/chemistry , Glutathione/metabolism , L-Iditol 2-Dehydrogenase/metabolism , L-Lactate Dehydrogenase/metabolism , Lung Diseases/chemically induced , Lung Diseases/pathology , Male , Mice , Quinolines/pharmacology , Reactive Oxygen Species/metabolism , Stereoisomerism , Trans-Activators/metabolism
15.
J Am Dent Assoc ; 140(2): 190-9; quiz 248-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19188416

ABSTRACT

BACKGROUND: The authors conducted a study to determine the effectiveness of two waste anesthetic gas-scavenging systems. They also evaluated one of the systems to determine the effect of work practices in controlling waste nitrous oxide (N2O). METHODS: The authors collected a minimum of 13 data sets in each phase of the study that included infrared thermography, digital videography and real-time air analysis for ambient concentrations of waste N2O. Surgeon 1, who had experience using both systems, used the Safe Sedate Dental Mask (Airgas, Radnor, Pa.) system (system I) in phase I and the Porter Nitrous Oxide Sedation System (Porter Instruments, Hatfield, Pa.) (system II) in phase II. Surgeon 2, who did not have experience using system I, used it in phase III. To evaluate each system's effectiveness, the authors collected N2O air concentration data from phases I and II and compared the data with the National Institute for Occupational Safety and Health Recommended Exposure Limit (NIOSH REL). They also compared phases I and III to determine the effect of work practices on the systems' effectiveness. RESULTS: Surgeon 1 controlled occupational exposure to N2O significantly better using system I than using system II. Mean N2O air concentration levels during phases I and II were 61.6 parts per million (ppm) and 225.6 ppm, respectively. Surgeon 2 did not achieve results comparable to those of surgeon 1 in phase I using system I. Infrared thermography and air concentration data suggested that key work practices and patient and surgical variables accounted for the different results obtained in phases I and III. CONCLUSIONS: Although neither system was able to control occupational exposure of N2O oxide below the NIOSH REL, system I met the American Conference of Governmental Industrial Hygienists threshold limit value of less than 50 ppm during an eight-hour day and performed significantly better than did system II. CLINICAL IMPLICATIONS: System I achieved maximal efficiency when combined with consistent best work practices.


Subject(s)
Anesthetics, Inhalation/analysis , Dental Equipment , Gas Scavengers , Nitrous Oxide/analysis , Occupational Exposure/prevention & control , Air Pollutants, Occupational/analysis , Humans , Infrared Rays , Oral Surgical Procedures , Practice Patterns, Dentists' , Spectrophotometry, Infrared , Thermography , Waste Products/analysis
16.
Am Heart J ; 157(2): 352-360.e2, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19185645

ABSTRACT

BACKGROUND: High-density lipoprotein cholesterol (HDL-C) levels are inversely associated with cardiovascular risk. Cholesteryl ester transfer protein inhibition is one strategy for increasing HDL-C. This study evaluated the lipid-altering efficacy and safety of the cholesteryl ester transfer protein inhibitor anacetrapib as monotherapy or coadministered with atorvastatin in patients with dyslipidemia. METHODS: A total of 589 patients with primary hypercholesterolemia or mixed hyperlipidemia (53.8% of the study population had low HDL-C) were randomized equally to one of 10 groups: 5 groups received background statin therapy of atorvastatin 20 mg and 5 did not, and each of these was randomized to placebo, anacetrapib 10, 40, 150, and 300 mg once daily for 8 weeks. An equal proportion of patients had triglycerides >150 mg/dL in each group. RESULTS: For placebo and anacetrapib monotherapy (10, 40, 150, and 300 mg), least squares mean percent changes from baseline to week 8 for low-density lipoprotein cholesterol (LDL-C) were 2%, -16%, -27%, -40%, and -39%, respectively, and for HDL-C were 4%, 44%, 86%, 139%, and 133%, respectively (P < .001 vs placebo for all doses). Coadministration of anacetrapib with atorvastatin produced significant incremental LDL-C reductions and similar HDL-C increases versus atorvastatin monotherapy. For both anacetrapib monotherapy and coadministration with atorvastatin, the LDL-C reductions were similar in patients with baseline triglyceride levels greater than and less than or equal to the median. Anacetrapib was well tolerated, and the incidence of adverse events was similar for placebo and all active treatment groups. There were no increases in systolic or diastolic blood pressure in any treatment arm. CONCLUSIONS: Anacetrapib, as monotherapy or coadministered with atorvastatin, produced significant reductions in LDL-C and increases in HDL-C; the net result of treatment with anacetrapib + atorvastatin was approximately 70% lowering of LDL-C and more than doubling of HDL-C. Anacetrapib was generally well tolerated with no discernable effect on blood pressure.


Subject(s)
Anticholesteremic Agents/therapeutic use , Dyslipidemias/drug therapy , Heptanoic Acids/therapeutic use , Oxazolidinones/therapeutic use , Pyrroles/therapeutic use , Aged , Atorvastatin , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Treatment Outcome
17.
Am J Vet Res ; 69(11): 1506-11, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18980434

ABSTRACT

OBJECTIVE: To apply the principle of sodium dilution to calculate the changes in the extracellular fluid (ECF) volume (ECFV) and intracellular fluid volume (ICFV) that occur during dehydration and rehydration in horses. ANIMALS: 8 healthy horses of various breeds. PROCEDURES: Horses were dehydrated over 4 hours by withholding water and administering furosemide. Saline (0.9% NaCl) solution was administered IV during the next 2 hours (20 mL/kg/h; total 40 mL/kg). Horses were monitored for an additional hour following IV fluid administration. Initial ECFV was determined by use of multifrequency bioelectrical impedance analysis, and serum sodium concentration was used to calculate total ECF sodium content. Sodium and fluid volume losses were monitored and calculated throughout the study and used to estimate changes in ECFV and ICFV during fluid balance alterations. RESULTS: Changes during dehydration and rehydration primarily occurred in the ECFV. The sodium dilution principle estimated an overexpansion of the ECFV beyond the volume of fluid administered, indicating a small contraction of the ICFV in response to fluid administration. Serum and urinary electrolyte changes were recorded and were consistent with those of previous reports. CONCLUSIONS AND CLINICAL RELEVANCE: The sodium dilution principle provided a simple method that can be used to estimate the changes in ECFV and ICFV that occur during fluid administration. Results suggested an overexpansion of the ECFV in response to IV saline solution administration. The sodium dilution principle requires further validation in healthy and clinically ill horses, which could provide clinical applications similar to those in other species.


Subject(s)
Dehydration/veterinary , Extracellular Fluid/metabolism , Fluid Therapy/veterinary , Horse Diseases/metabolism , Animals , Dehydration/metabolism , Dehydration/therapy , Electrolytes/blood , Electrolytes/urine , Horse Diseases/therapy , Horses , Indicator Dilution Techniques/veterinary , Sodium
18.
Toxicol Lett ; 183(1-3): 28-35, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18926891

ABSTRACT

Styrene, widely used in manufacturing, has both acute and chronic effects in humans. In mice, styrene is both hepato- and pneumo-toxic and causes lung tumors. The primary site for styrene metabolism and its effects in mouse lung is the Clara cell, which secretes Clara cell 10kDa protein (CC10) and surfactant protein A (SPA). Both play important roles in host defenses and inflammation prevention. The mode of action for styrene-induced lung tumor formation has yet to be elicited, yet one possibility relates to oxidative stress and decreased CC10 levels. CC10 mRNA and protein expression were measured in isolated Clara cells 3, 12, and 24h following in vivo administration of styrene (600mg/kg i.p.) or its metabolites [R-, S-, racemic styrene oxide (SO) (300mg/kg i.p.), 4-vinylphenol (100mg/kg i.p.)]. The largest decreases in CC10 mRNA expression were seen with R-SO and racemic SO at 24h. To determine if rebound effects would be seen, CC10 mRNA and protein expression were determined 48, 120, and 240h following styrene and R-SO administration. The CC10 protein level did not reach its lowest point to correlate with mRNA expression until 120h after R-SO administration. Styrene exposure caused a significant decrease in CC10 protein after 24h, rebounding through 240h. SPA protein expression showed little change from control levels, indicating a more specific effect on CC10 in the Clara cell by styrene and its metabolites. These studies demonstrate that acute changes in lung CC10 protein and mRNA expression do occur following in vivo treatment with styrene and its metabolites. These changes may be early indicators for a potential mechanism for lung tumor formation in mice as it relates to oxidative stress and the possibility deserves further study.


Subject(s)
Epoxy Compounds/toxicity , Lung/drug effects , Phenols/toxicity , Uteroglobin/metabolism , Age Factors , Animals , Blotting, Western , Cells, Cultured , Epoxy Compounds/administration & dosage , Epoxy Compounds/metabolism , Gene Expression/drug effects , Injections, Intraperitoneal , Lung/cytology , Lung/metabolism , Male , Mice , Phenols/administration & dosage , Phenols/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Uteroglobin/genetics
19.
Toxicology ; 254(1-2): 1-10, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18951947

ABSTRACT

Styrene is widely used with significant human exposure, particularly in the reinforced plastics industry. In mice it is both hepatotoxic and pneumotoxic, and this toxicity is generally thought to be associated with its metabolism to styrene oxide. Styrene causes lung tumors in mice but not in rats. The question is how the tumorigenic effect in mouse lung may relate to the human. This review examines the comparison of the metabolic activation rates (1) between the liver and lung and (2) for the lung, between the rodent and human. Emphasis is placed on the specific cytochromes P450 present in the lungs of humans and what role they might play in the bioactivation of styrene and other compounds. In general, pulmonary metabolism is very slow compared to hepatic metabolism. Furthermore, metabolic rates in humans are slow compared to those in rats and mice. There is a wide difference in what specific cytochromes P450 investigators have reported as being present in human lung which makes comparisons, both inter-species and inter-organ, difficult. The general low activity for cytochrome P450 activity in the lung, especially for CYP2F1, the human homolog for CYP2F2 which has been identified in mice as being primarily responsible for styrene metabolism, argues against the hypothesis that human lung would produce enough styrene oxide to damage pulmonary epithelial cells leading to cell death, increased cell replication and ultimately tumorigenicity, the presumed mode of action for styrene in the production of the mouse lung tumors.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Lung Neoplasms/chemically induced , Lung/enzymology , Styrene/toxicity , Carcinogens/toxicity , Humans , Inhalation Exposure , Microsomes/metabolism , Styrene/pharmacokinetics
20.
Scanning ; 30(2): 78-86, 2008.
Article in English | MEDLINE | ID: mdl-18241039

ABSTRACT

Latex films have a tendency to "blush" when exposed to water. The swelling of trapped hydrophilic material, which results in pockets with different refractive indices, is proposed as a possible model for blushing. A pressure-sensitive latex was developed on the basis of this model. The blush was improved by the choice of the surfactant and control of the particle diameter. The resulting latex has an unusual particle growth behavior during polymerization. The particle size distribution (PSD) was characterized by dynamic light scattering (DLS), electron microscopy (SEM/TEM) and atomic force microscopy (AFM). The data are in a good agreement with our proposed growth model.

SELECTION OF CITATIONS
SEARCH DETAIL
...