Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Genes (Basel) ; 15(1)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38254988

ABSTRACT

This clinical study examined the influence of SLCO1B1 c.521T>C (rs4149056) on plasma atorvastatin concentrations in pediatric hypercholesterolemia. The participants (8-21 years), including heterozygous (c.521T/C, n = 13), homozygous (c.521C/C, n = 2) and controls (c.521T/T, n = 13), completed a single-oral-dose pharmacokinetic study. Similar to in adults, the atorvastatin (AVA) area-under-concentration-time curve from 0 to 24 h (AUC0-24) was 1.7-fold and 2.8-fold higher in participants with c.521T/C and c.521C/C compared to the c.521T/T participants, respectively. The inter-individual variability in AVA exposure within these genotype groups ranged from 2.3 to 4.8-fold, indicating that additional factors contribute to the inter-individual variability in the AVA dose-exposure relationship. A multivariate model reinforced the SLCO1B1 c.521T>C variant as the central factor contributing to AVA systemic exposure in this pediatric cohort, accounting for ~65% of the variability in AVA AUC0-24. Furthermore, lower AVA lactone concentrations in participants with increased body mass index contributed to higher exposure within the c.521T/T and c.521T/C genotype groups. Collectively, these factors contributing to higher systemic exposure could increase the risk of toxicity and should be accounted for when individualizing the dosing of atorvastatin in eligible pediatric patients.


Subject(s)
Hypercholesterolemia , Adult , Humans , Child , Hypercholesterolemia/drug therapy , Hypercholesterolemia/genetics , Atorvastatin/therapeutic use , Genotype , Heterozygote , Genetic Variation , Liver-Specific Organic Anion Transporter 1/genetics
2.
Chem Res Toxicol ; 35(10): 1634-1635, 2022 Oct 17.
Article in English | MEDLINE | ID: mdl-35536050
3.
Cell Death Dis ; 12(6): 562, 2021 05 31.
Article in English | MEDLINE | ID: mdl-34059639

ABSTRACT

Ciclopirox (CPX) is an FDA-approved topical antifungal agent that has demonstrated preclinical anticancer activity in a number of solid and hematologic malignancies. Its clinical utility as an oral anticancer agent, however, is limited by poor oral bioavailability and gastrointestinal toxicity. Fosciclopirox, the phosphoryloxymethyl ester of CPX (Ciclopirox Prodrug, CPX-POM), selectively delivers the active metabolite, CPX, to the entire urinary tract following parenteral administration. We characterized the activity of CPX-POM and its major metabolites in in vitro and in vivo preclinical models of high-grade urothelial cancer. CPX inhibited cell proliferation, clonogenicity and spheroid formation, and increased cell cycle arrest at S and G0/G1 phases. Mechanistically, CPX suppressed activation of Notch signaling. Molecular modeling and cellular thermal shift assays demonstrated CPX binding to γ-secretase complex proteins Presenilin 1 and Nicastrin, which are essential for Notch activation. To establish in vivo preclinical proof of principle, we tested fosciclopirox in the validated N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) mouse bladder cancer model. Once-daily intraperitoneal administration of CPX-POM for four weeks at doses of 235 mg/kg and 470 mg/kg significantly decreased bladder weight, a surrogate for tumor volume, and resulted in a migration to lower stage tumors in CPX-POM treated animals. This was coupled with a reduction in the proliferation index. Additionally, there was a reduction in Presenilin 1 and Hes-1 expression in the bladder tissues of CPX-POM treated animals. Following the completion of the first-in-human Phase 1 trial (NCT03348514), the pharmacologic activity of fosciclopirox is currently being characterized in a Phase 1 expansion cohort study of muscle-invasive bladder cancer patients scheduled for cystectomy (NCT04608045) as well as a Phase 2 trial of newly diagnosed and recurrent urothelial cancer patients scheduled for transurethral resection of bladder tumors (NCT04525131).


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Antifungal Agents/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Ciclopirox/therapeutic use , Antifungal Agents/pharmacology , Ciclopirox/pharmacology , Humans , Neoplasm Grading
4.
Clin Cancer Res ; 27(14): 3896-3904, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33602685

ABSTRACT

PURPOSE: PIK3CA mutations are common in breast cancer and promote tumor progression and treatment resistance. We conducted a phase I/II trial of alpelisib (α-specific PI3K inhibitor) plus nab-paclitaxel in patients with HER2-negative metastatic breast cancer (MBC). PATIENTS AND METHODS: Eligible patients had HER2-negative MBC with any number of prior chemotherapies. Phase I was 3+3 dose-escalation design with three dose levels of alpelisib (250, 300, and 350 mg) daily plus nab-paclitaxel 100 mg/m2 administered on days 1, 8, and 15 every 28 days. Phase II was according to Simon's two-stage design. PIK3CA mutations in tumor/circulating tumor DNA (ctDNA) were assessed. Primary endpoints were recommended phase II dose (RP2D) and objective response rate (ORR). Additional endpoints included safety, pharmacokinetics, progression-free survival (PFS), and association of PIK3CA mutation with outcomes. RESULTS: A total of 43 patients were enrolled (phase I, n = 13 and phase II, n = 30). A total of 84% had visceral disease and 84% had prior taxane. No dose-limiting toxicities occurred in phase I. RP2D was alpelisib 350 mg daily plus nab-paclitaxel 100 mg/m2 on days 1, 8, and 15. Hyperglycemia (grade 3, 26% and grade 4, 0%), neutropenia (grade 3, 23% and grade 4, 7%), diarrhea (grade 3, 5% and grade 4, 0%), and rash (grade 3, 7% and grade 4, 0%) were the most common adverse events. Among 42 evaluable patients, ORR was 59% (complete response, 7% and partial response, 52%), 21% of whom had response lasting >12 months; median PFS was 8.7 months. A total of 40% of patients demonstrated tumor and/or ctDNA PIK3CA mutation; patients with tumor/ctDNA mutation demonstrated better PFS compared with those without mutation (11.9 vs. 7.5 months; HR, 0.44; P = 0.027). Patients with normal metabolic status had longer PFS compared with prediabetic/diabetic patients (12 vs. 7.5 months; P = 0.014). No pharmacokinetics interactions were detected. CONCLUSIONS: The alpelisib plus nab-paclitaxel combination was well tolerated and shows encouraging efficacy, especially in patients with PIK3CA-mutated tumor/ctDNA. The impact of metabolic status on response to this combination merits further investigation.


Subject(s)
Albumins/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Mutation , Paclitaxel/administration & dosage , Thiazoles/administration & dosage , Adult , Aged , Biomarkers, Tumor/blood , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Drug Combinations , Female , Humans , Middle Aged , Neoplasm Metastasis , Receptor, ErbB-2/analysis
5.
Alzheimers Dement ; 17(1): 7-17, 2021 01.
Article in English | MEDLINE | ID: mdl-32715609

ABSTRACT

INTRODUCTION: Brain bioenergetics are defective in Alzheimer's disease (AD). Preclinical studies find oxaloacetate (OAA) enhances bioenergetics, but human safety and target engagement data are lacking. METHODS: We orally administered 500 or 1000 mg OAA, twice daily for 1 month, to AD participants (n = 15 each group) and monitored safety and tolerability. To assess brain metabolism engagement, we performed fluorodeoxyglucose positron emission tomography (FDG PET) and magnetic resonance spectroscopy before and after the intervention. We also assessed pharmacokinetics and cognitive performance. RESULTS: Both doses were safe and tolerated. Compared to the lower dose, the higher dose benefited FDG PET glucose uptake across multiple brain regions (P < .05), and the higher dose increased parietal and frontoparietal glutathione (P < .05). We did not demonstrate consistent blood level changes and cognitive scores did not improve. CONCLUSIONS: 1000 mg OAA, taken twice daily for 1 month, is safe in AD patients and engages brain energy metabolism.


Subject(s)
Alzheimer Disease/drug therapy , Oxaloacetic Acid/administration & dosage , Oxaloacetic Acid/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Brain/drug effects , Brain/metabolism , Cognition/drug effects , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Female , Fluorodeoxyglucose F18 , Glucose/metabolism , Glutathione/metabolism , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Middle Aged , Neuropsychological Tests , Oxaloacetic Acid/adverse effects , Positron-Emission Tomography , Radiopharmaceuticals
6.
J Pharmacol Exp Ther ; 370(2): 148-159, 2019 08.
Article in English | MEDLINE | ID: mdl-31113837

ABSTRACT

Pharmacokinetic studies in rats and dogs were performed to characterize the in vivo performance of a novel prodrug, fosciclopirox. Ciclopirox olamine (CPX-O) is a marketed topical antifungal agent with demonstrated in vitro and in vivo preclinical anticancer activity in several solid tumor and hematologic malignancies. The oral route of administration for CPX-O is not feasible due to low bioavailability and dose-limiting gastrointestinal toxicities. To enable parenteral administration, the phosphoryl-oxymethyl ester of ciclopirox (CPX), fosciclopirox (CPX-POM), was synthesized and formulated as an injectable drug product. In rats and dogs, intravenous CPX-POM is rapidly and completely metabolized to its active metabolite, CPX. The bioavailability of the active metabolite is complete following CPX-POM administration. CPX and its inactive metabolite, ciclopirox glucuronide (CPX-G), are excreted in urine, resulting in delivery of drug to the entire urinary tract. The absolute bioavailability of CPX following subcutaneous administration of CPX-POM is excellent in rats and dogs, demonstrating the feasibility of this route of administration. These studies confirmed the oral bioavailability of CPX-O is quite low in rats and dogs compared with intravenous CPX-POM. Given its broad-spectrum anticancer activity in several solid tumor and hematologic cancers and renal elimination, CPX-POM is being developed for the treatment of urothelial cancer. The safety, dose tolerance, pharmacokinetics, and pharmacodynamics of intravenous CPX-POM are currently being characterized in a United States multicenter first-in-human Phase 1 clinical trial in patients with advanced solid tumors (NCT03348514).


Subject(s)
Ciclopirox/metabolism , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Urologic Neoplasms/drug therapy , Urologic Neoplasms/pathology , Urothelium/drug effects , Animals , Biological Availability , Dogs , Male , Prodrugs/metabolism , Prodrugs/therapeutic use , Rats
7.
Sci Rep ; 7(1): 17188, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215048

ABSTRACT

Pancreatic cancer is among the most lethal cancers with poorly tolerated treatments. There is increasing interest in using high-dose intravenous ascorbate (IVC) in treating this disease partially because of its low toxicity. IVC bypasses bioavailability barriers of oral ingestion, provides pharmacological concentrations in tissues, and exhibits selective cytotoxic effects in cancer cells through peroxide formation. Here, we further revealed its anti-pancreatic cancer mechanisms and conducted a phase I/IIa study to investigate pharmacokinetic interaction between IVC and gemcitabine. Pharmacological ascorbate induced cell death in pancreatic cancer cells with diverse mutational backgrounds. Pharmacological ascorbate depleted cellular NAD+ preferentially in cancer cells versus normal cells, leading to depletion of ATP and robustly increased α-tubulin acetylation in cancer cells. While ATP depletion led to cell death, over-acetylated tubulin led to inhibition of motility and mitosis. Collagen was increased, and cancer cell epithelial-mesenchymal transition (EMT) was inhibited, accompanied with inhibition in metastasis. IVC was safe in patients and showed the possibility to prolong patient survival. There was no interference to gemcitabine pharmacokinetics by IVC administration. Taken together, these data revealed a multi-targeting mechanism of pharmacological ascorbate's anti-cancer action, with minimal toxicity, and provided guidance to design larger definitive trials testing efficacy of IVC in treating advanced pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Ascorbic Acid/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Follow-Up Studies , Humans , Infusions, Parenteral , Mice , Mice, Nude , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Prognosis , Prospective Studies , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
8.
Curr Protoc Pharmacol ; 75: 7.6.1-7.6.12, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27960029

ABSTRACT

Determination of drug or drug metabolite concentrations in biological samples, particularly in serum or plasma, is fundamental to describing the relationships between administered dose, route of administration, and time after dose for achieving the optimal clinical response. While a well-characterized, accurate analytical method is needed to define these parameters, it must also be established that the analyte concentration in the sample at the time of analysis is identical to the concentration at sample acquisition. This is necessitated by the fact that drugs and their metabolites are susceptible to degradation in samples due to metabolism or to physical and chemical processes, resulting in a lower measured concentration than was in the original sample. Careful examination of analyte stability during processing and storage and, if necessary, adjustment of procedures and conditions to maximize stability, are a critical component of method validation to ensure the accuracy of the data. The protocols provided in this unit address the stability of the analytes in whole blood and blood-derived samples prior to sample preparation for analysis. Issues addressed include sample acquisition, processing of whole blood, and storage of blood-derived samples. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Drug Stability , Pharmaceutical Preparations , Chromatography, Liquid/methods , Humans , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/isolation & purification , Pharmaceutical Preparations/metabolism , Plasma/chemistry , Plasma/metabolism , Tandem Mass Spectrometry/methods
9.
Cancer Med ; 5(11): 3031-3040, 2016 11.
Article in English | MEDLINE | ID: mdl-27734609

ABSTRACT

Acute myeloid leukemia (AML) cells meet the higher energy, metabolic, and signaling demands of the cell by increasing mitochondrial biogenesis and mitochondrial protein translation. Blocking mitochondrial protein synthesis through genetic and chemical approaches kills human AML cells at all stages of development in vitro and in vivo. Tigecycline is an antimicrobial that we found inhibits mitochondrial protein synthesis in AML cells. Therefore, we conducted a phase 1 dose-escalation study of tigecycline administered intravenously daily 5 of 7 days for 2 weeks to patients with AML. A total of 27 adult patients with relapsed and refractory AML were enrolled in this study with 42 cycles being administered over seven dose levels (50-350 mg/day). Two patients experienced DLTs related to tigecycline at the 350 mg/day level resulting in a maximal tolerated dose of tigecycline of 300 mg as a once daily infusion. Pharmacokinetic experiments showed that tigecycline had a markedly shorter half-life in these patients than reported for noncancer patients. No significant pharmacodynamic changes or clinical responses were observed. Thus, we have defined the safety of once daily tigecycline in patients with refractory AML. Future studies should focus on schedules of the drug that permit more sustained target inhibition.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Minocycline/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Drug Monitoring , Drug Resistance, Neoplasm , Female , Humans , Infusions, Intravenous , Leukemia, Myeloid, Acute/diagnosis , Male , Maximum Tolerated Dose , Middle Aged , Minocycline/administration & dosage , Minocycline/adverse effects , Minocycline/pharmacokinetics , Mitochondria/drug effects , Mitochondria/metabolism , Recurrence , Retreatment , Risk Assessment , Tigecycline , Treatment Outcome
10.
BBA Clin ; 5: 120-3, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27051598

ABSTRACT

Bioenergetics and bioenergetic-related functions are altered in Alzheimer's disease (AD) subjects. These alterations represent therapeutic targets and provide an underlying rationale for modifying brain bioenergetics in AD-affected persons. Preclinical studies in cultured cells and mice found that administering oxaloacetate (OAA), a Krebs cycle and gluconeogenesis intermediate, enhanced bioenergetic fluxes and upregulated some brain bioenergetic infrastructure-related parameters. We therefore conducted a study to provide initial data on the tolerability and pharmacokinetics of OAA in AD subjects. Six AD subjects received OAA 100 mg capsules twice a day for one month. The intervention was well-tolerated. Blood level measurements following ingestion of a 100 mg OAA capsule showed modest increases in OAA concentrations, but pharmacokinetic analyses were complicated by relatively high amounts of endogenous OAA. We conclude that OAA 100 mg capsules twice per day for one month are safe in AD subjects but do not result in a consistent and clear increase in the OAA blood level, thus necessitating future clinical studies to evaluate higher doses.

11.
Cancer Chemother Pharmacol ; 75(5): 1075-87, 2015 May.
Article in English | MEDLINE | ID: mdl-25898813

ABSTRACT

PURPOSE: This multicenter, open-label, dose-escalating, phase I study evaluated the safety, tolerability, pharmacokinetics and preliminary tumor response of a nanoparticulate formulation of paclitaxel (Nanotax®) administered intraperitoneally for multiple treatment cycles in patients with solid tumors predominantly confined to the peritoneal cavity for whom no other curative systemic therapy treatment options were available. METHODS: Twenty-one patients with peritoneal malignancies received Nanotax® in a modified dose-escalation approach utilizing an accelerated titration method. All patients enrolled had previously received chemotherapeutics and undergone surgical procedures, including 33 % with optimal debulking. Six doses (50-275 mg/m(2)) of Cremophor-free Nanotax® were administered intraperitoneally for one to six cycles (every 28 days). RESULTS: Intraperitoneal (IP) administration of Nanotax® did not lead to increases in toxicity over that typically associated with intravenous (IV) paclitaxel. No patient reported ≥Grade 2 neutropenia and/or ≥Grade 3 neurologic toxicities. Grade 3 thrombocytopenia unlikely related to study medication occurred in one patient. The peritoneal concentration-time profile of paclitaxel rose during the 2 days after dosing to peritoneal fluid concentrations 450-2900 times greater than peak plasma drug concentrations and remained elevated through the entire dose cycle. Best response assessments were made in 16/21 patients: Four patients were assessed as stable or had no response and twelve patients had increasing disease. Five of 21 patients with advanced cancers survived longer than 400 days after initiation of Nanotax® IP treatment. CONCLUSIONS: Compared to IV paclitaxel administration, Cremophor-free IP administration of Nanotax® provides higher and prolonged peritoneal paclitaxel levels with minimal systemic exposure and reduced toxicity.


Subject(s)
Nanoparticles/administration & dosage , Nanoparticles/adverse effects , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Peritoneal Neoplasms/drug therapy , Adult , Aged , Dose-Response Relationship, Drug , Female , Humans , Infusions, Parenteral , Male , Middle Aged , Nanoparticles/metabolism , Paclitaxel/pharmacokinetics , Peritoneal Neoplasms/metabolism
12.
Environ Manage ; 55(6): 1366-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25832342

ABSTRACT

We are using bobcats (Lynx rufus) as a model organism to examine how roads affect the abundance, distribution, and genetic structure of a wide-ranging carnivore. First, we compared the distribution of bobcat-vehicle collisions to road density and then estimated collision probabilities for specific landscapes using a moving window with road-specific traffic volume. Next, we obtained incidental observations of bobcats from the public, camera-trap detections, and locations of bobcats equipped with GPS collars to examine habitat selection. These data were used to generate a cost-surface map to investigate potential barrier effects of roads. Finally, we have begun an examination of genetic structure of bobcat populations in relation to major road networks. Distribution of vehicle-killed bobcats was correlated with road density, especially state and interstate highways. Collision models suggested that some regions may function as demographic sinks. Simulated movements in the context of the cost-surface map indicated that some major roads may be barriers. These patterns were supported by the genetic structure of bobcats. The sharpest divisions among genetically distinct demes occurred along natural barriers (mountains and large lakes) and in road-dense regions. In conclusion, our study has demonstrated the utility of using bobcats as a model organism to understand the variety of threats that roads pose to a wide-ranging species. Bobcats may also be useful as one of a group of focal species while developing approaches to maintain existing connectivity or mitigate the negative effects of roads.


Subject(s)
Accidents, Traffic/prevention & control , Ecosystem , Environment Design , Lynx/growth & development , Models, Biological , Animals , Genetics, Population , Genotype , Lynx/genetics , New Hampshire , Risk Factors , Urbanization
13.
J Child Adolesc Psychopharmacol ; 25(2): 150-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25782098

ABSTRACT

OBJECTIVES: Our clinical experience with low dose loxapine (5-15 mg/day) suggests promising efficacy and safety for irritability in autism spectrum disorders (ASD). We studied low dose loxapine prospectively in adolescents and adults with ASD and irritability. Additionally, we measured loxapine and metabolite concentrations, and brain-derived neurotrophic factor (BDNF) as a biomarker of neuromodulation. METHODS: We performed a 12 week open trial of add-on loxapine in subjects, ages 13-65 years, diagnosed with ASD, and Aberrant Behavior Checklist-Irritability (ABC-I) subscale scores >14. Loxapine was dosed flexibly up to 15 mg daily, starting with 5 mg on alternate days. From weeks 1 to 6, other psychoactive medications were tapered if possible; from weeks 6 to 12, all medication doses were held stable. The primary outcome was the Clinical Global Impressions-Improvement subscale (CGI-I), ratings of Much Improved or Very Much Improved. Secondary outcomes were the ABC-I, Repetitive Behavior Scale-Revised, and Schalock Quality of Life scale. Serum BDNF and loxapine and metabolite concentrations were assayed. BDNF rs6265 was genotyped. RESULTS: Sixteen subjects were enrolled; 12 completed all visits. Median age was 18 years (range 13-39). Median final loxapine dose was 7.5 mg/day (2.5-15). All 14 subjects (100%) with data at week 12 were rated as Much Improved on CGI-I at 12 weeks. Mean change on ABC-I at 12 weeks was -31%, p=0.01. Mean body mass index (BMI)-Z decreased between weeks 6 and 12, p=0.03. Side effects were minimal, and prolactin elevation occurred in only one subject. BDNF concentrations measured in 11 subjects increased significantly (p=0.04). Subjects with AG genotype for BDNF rs6265 required a lower dose of loxapine at study end, but had similar behavioral and BDNF concentration changes as the GG genotype. CONCLUSIONS: Low dose loxapine shows promise as a repurposed drug for irritability in ASD. Loxapine effects on BDNF warrant further study.


Subject(s)
Antipsychotic Agents/administration & dosage , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/psychology , Irritable Mood/drug effects , Loxapine/administration & dosage , Adolescent , Adult , Autism Spectrum Disorder/blood , Brain-Derived Neurotrophic Factor/blood , Drug Therapy, Combination , Female , Humans , Male , Prospective Studies , Psychiatric Status Rating Scales , Treatment Outcome , Young Adult
14.
PLoS One ; 9(7): e102165, 2014.
Article in English | MEDLINE | ID: mdl-25020133

ABSTRACT

Although increased serum histamine levels and H1R expression in the plaque are seen in atherosclerosis, it is not known whether H1R activation is a causative factor in the development of the disease, or is a host defense response to atherogenic signals. In order to elucidate how pharmacological inhibition of histamine receptor 1 (H1R) signaling affects atherogenesis, we administered either cetirizine (1 and 4 mg/kg. b.w) or fexofenadine (10 and 40 mg/kg. b.w) to ApoE-/- mice maintained on a high fat diet for three months. Mice ingesting a low dose of cetirizine or fexofenadine had significantly higher plaque coverage in the aorta and cross-sectional lesion area at the aortic root. Surprisingly, the higher doses of cetirizine or fexofenadine did not enhance atherosclerotic lesion coverage over the controls. The low dose of fexofenadine, but not cetirizine, increased serum LDL cholesterol. Interestingly, the expression of iNOS and eNOS mRNA was increased in aortas of mice on high doses of cetirizine or fexofenadine. This may be a compensatory nitric oxide (NO)-mediated vasodilatory mechanism that accounts for the lack of increase in the progression of atherosclerosis. Although the administration of cetirizine did not alter blood pressure between the groups, there was a positive correlation between blood pressure and lesion/media ratio at the aortic root in mice receiving the low dose of cetirizine. However, this association was not observed in mice treated with the high dose of cetirizine or either doses of fexofenadine. The macrophages or T lymphocytes densities were not altered by low doses of H1-antihistamines, whereas, high doses decreased the number of macrophages but not T lymphocytes. The number of mast cells was decreased only in mice treated with low dose of fexofenadine. These results demonstrate that chronic ingestion of low therapeutic doses of cetirizine or fexofenadine enhance progression of atherosclerosis.


Subject(s)
Atherosclerosis/chemically induced , Atherosclerosis/pathology , Cetirizine/adverse effects , Histamine H1 Antagonists/adverse effects , Terfenadine/analogs & derivatives , Analysis of Variance , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Blood Chemical Analysis , CD36 Antigens/metabolism , Cetirizine/blood , Cetirizine/pharmacology , Cholesterol, LDL/blood , Diet, High-Fat , Disease Progression , Fluorescent Antibody Technique , Histamine H1 Antagonists/pharmacology , Male , Mast Cells/metabolism , Mice , Mice, Knockout , Nitric Oxide Synthase/metabolism , Signal Transduction/drug effects , Terfenadine/adverse effects , Terfenadine/blood , Terfenadine/pharmacology
15.
Am J Physiol Gastrointest Liver Physiol ; 307(2): G219-28, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24852568

ABSTRACT

We examined the effects of two over-the-counter H1-antihistamines on the progression of fatty liver disease in male C57Bl/6 wild-type and apolipoprotein E (ApoE)-/- mice. Mice were fed a high-fat diet (HFD) for 3 mo, together with administration of either cetirizine (4 mg/kg body wt) or fexofenadine (40 mg/kg body wt) in drinking water. Antihistamine treatments increased body weight gain, gonadal fat deposition, liver weight, and hepatic steatosis in wild-type mice but not in ApoE-/- mice. Lobular inflammation, acute inflammation, and necrosis were not affected by H1-antihistamines in either genotype. Serum biomarkers of liver injury tended to increase in antihistamine-treated wild-type mice. Serum level of glucose was increased by fexofenadine, whereas lipase was increased by cetirizine. H1-antihistamines reduced the mRNA expression of ApoE and carbohydrate response element-binding protein in wild-type mice, without altering the mRNA expression of sterol regulatory element-binding protein 1c, fatty acid synthase, or ApoB100, in either genotype. Fexofenadine increased both triglycerides and cholesterol ester, whereas cetirizine increased only cholesterol ester in liver, with a concomitant decrease in serum triglycerides by both antihistamines in wild-type mice. Antihistamines increased hepatic levels of conjugated bile acids in wild-type mice, with the effect being significant in fexofenadine-treated animals. The increase was associated with changes in the expression of organic anion transport polypeptide 1b2 and bile salt export pump. These results suggest that H1-antihistamines increase the progression of fatty liver disease in wild-type mice, and there seems to be an association between the severity of disease, presence of ApoE, and increase in hepatic bile acid levels.


Subject(s)
Apolipoproteins E/deficiency , Cetirizine/toxicity , Diet, High-Fat , Fatty Liver/chemically induced , Histamine H1 Antagonists/toxicity , Liver/drug effects , Terfenadine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoproteins E/genetics , Bile Acids and Salts/metabolism , Biomarkers/blood , Cholesterol Esters/metabolism , Disease Models, Animal , Fatty Liver/blood , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression Regulation , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/metabolism , Liver/pathology , Liver-Specific Organic Anion Transporter 1 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Severity of Illness Index , Terfenadine/toxicity , Triglycerides/metabolism
16.
Environ Int ; 66: 174-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24607502

ABSTRACT

Temuco is one of the most highly wood smoke polluted cities in Chile; however, there is scarce evidence of respiratory morbidity due to fine particulate matter. We aimed to estimate the relationship between daily concentration of ultrafine particles (UFP), with an aerodynamic diameter ≤ 0.1 µm, and outpatient visits for respiratory illness at medical care centers of Temuco, Chile, from August the 20th, 2009 to June the 30th, 2011. The Air Pollution Health Effects European Approach (APHEA2) protocol was followed, and a multivariate semi-parametric Poisson regression model was fitted with GAM techniques using R-Project statistical package; controlling for trend, seasonality, and confounders. The daily UFP were measured by a MOUDI NR-110 sampler. We found that results of the statistical analyses show significant associations between UFP and respiratory outpatient visits, with the elderly (population ≥ 65 years), being the group that presented the greatest risk. An interquartile increase of 4.73 µg/m(3) in UFP (lag 5 days) was associated with respiratory outpatient visits with a relative risk (RR) of 1.1458 [95% CI (1.0497-1.2507)] for the elderly. These results show novel findings regarding the relevance of daily UFP concentrations and health risk, especially for susceptible population in a wood smoke polluted city.


Subject(s)
Environmental Exposure , Particle Size , Particulate Matter/toxicity , Respiratory System/drug effects , Respiratory Tract Diseases/epidemiology , Smoke/adverse effects , Wood/chemistry , Adolescent , Adult , Aged , Child , Child, Preschool , Chile/epidemiology , Demography , Humans , Middle Aged , Regression Analysis , Risk , Time , Young Adult
17.
Am J Hematol ; 89(4): 363-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24273151

ABSTRACT

The antimycotic ciclopirox olamine is an intracellular iron chelator that has anticancer activity in vitro and in vivo. We developed an oral formulation of ciclopirox olamine and conducted the first-in-human phase I study of this drug in patients with relapsed or refractory hematologic malignancies (Trial registration ID: NCT00990587). Patients were treated with 5-80 mg/m² oral ciclopirox olamine once daily for five days in 21-day treatment cycles. Pharmacokinetic and pharmacodynamic companion studies were performed in a subset of patients. Following definition of the half-life of ciclopirox olamine, an additional cohort was enrolled and treated with 80 mg/m² ciclopirox olamine four times daily. Adverse events and clinical response were monitored throughout the trial. Twenty-three patients received study treatment. Ciclopirox was rapidly absorbed and cleared with a short half-life. Plasma concentrations of an inactive ciclopirox glucuronide metabolite were greater than those of ciclopirox. Repression of survivin expression was observed in peripheral blood cells isolated from patients treated once daily with ciclopirox olamine at doses greater than 10 mg/m², demonstrating biological activity of the drug. Dose-limiting gastrointestinal toxicities were observed in patients receiving 80 mg/m² four times daily, and no dose limiting toxicity was observed at 40 mg/m² once daily. Hematologic improvement was observed in two patients. Once-daily dosing of oral ciclopirox olamine was well tolerated in patients with relapsed or refractory hematologic malignancies, and further optimization of dosing regimens is warranted in this patient population.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematologic Neoplasms/drug therapy , Iron Chelating Agents/therapeutic use , Pyridones/therapeutic use , Salvage Therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Ciclopirox , Female , Gastrointestinal Diseases/chemically induced , Gene Expression Regulation, Neoplastic/drug effects , Half-Life , Hematologic Neoplasms/blood , Hematologic Neoplasms/pathology , Humans , Inactivation, Metabolic , Inhibitor of Apoptosis Proteins/genetics , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacokinetics , Male , Middle Aged , Neoplasm Proteins/genetics , Pyridones/adverse effects , Pyridones/blood , Pyridones/pharmacokinetics , RNA, Messenger/blood , RNA, Neoplasm/blood , Survivin , Treatment Outcome
19.
Nicotine Tob Res ; 14(6): 688-93, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22318754

ABSTRACT

INTRODUCTION: Despite the widespread use of mentholated cigarettes, lower cessation rates, and disproportionately high smoking-related morbidity among Blacks, the possible role of menthol in smokers' response to pharmacotherapy has not been well-studied. This study examined the effects of menthol on the pharmacokinetic (PK) profiles of bupropion and its principal metabolites, hydroxybupropion, threohydrobupropion, and erythrohydrobupropion among Black smokers. METHODS: After a 7-day placebo run-in period, participants received 150 mg bid sustained-release bupropion for 20-25 days. Blood samples were drawn for PK analysis on 2 occasions, 10-15 days after the commencement of bupropion while participants were still smoking (smoking phase) and at days 20-25 when they were asked not to smoke (nonsmoking phase). RESULTS: 18 smokers of nonmenthol cigarettes and 23 smokers of menthol cigarettes were enrolled in this study. No differences were found by menthol smoking status in the Cmax and area under the plasma concentration versus time curve (AUC) of bupropion and its metabolites in the smoking or nonsmoking phases. However, among menthol smokers, the AUC ratios of metabolite/bupropion were lower in the nonsmoking phase compared with the smoking phase (hydro/bup = 31.49 ± 18.84 vs. 22.95 ± 13.27, p = .04; erythro/bup = 1.99 ± 1.02 vs. 1.76 ± 0.75, p = .016; threo/bup = 11.77 ± 8.90 vs. 10.44 ± 5.63, p = .034). No significant differences were found in the metabolite/bup ratios between smoking and nonsmoking conditions among nonmenthol smokers. CONCLUSIONS: We did not find a significant effect of menthol compared with nonmenthol cigarette smoking on the PKs of bupropion and metabolites at steady state. More research is needed to advance the understanding of mechanisms underlying disparities in smoking cessation outcomes related to smoking of menthol cigarettes.


Subject(s)
Black People , Bupropion/pharmacokinetics , Menthol/pharmacology , Smoking Cessation/ethnology , Smoking/ethnology , Adolescent , Area Under Curve , Body Mass Index , Bupropion/analogs & derivatives , Bupropion/blood , Cotinine/analysis , Female , Humans , Male , Menthol/administration & dosage , Smoking Cessation/methods , Smoking Prevention , Time Factors , Tobacco Use Cessation Devices/statistics & numerical data , Young Adult
20.
Curr Pharm Biotechnol ; 13(1): 173-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21466430

ABSTRACT

Cancer is one of the leading causes of death in the United States and accounts for approximately 8 million deaths per year worldwide. Although there is an increasing number of therapeutic options available for patients with cancer, their efficacy is time-limited and non-curative. Approximately 50-60% cancer patients in the United States utilize agents derived from different parts of plants or nutrients (complementary and alternative medicine), exclusively or concurrently with traditional therapeutic regime such as chemotherapy and/or radiation therapy. The need for new drugs has prompted studies evaluating possible anti-cancer agents in fruits, vegetables, herbs and spices. Saffron, a spice and a food colorant present in the dry stigmas of the plant Crocus sativus L., has been used as an herbal remedy for various ailments including cancer by the ancient Arabian, Indian and Chinese cultures. Crocetin, an important carotenoid constituent of saffron, has shown significant potential as an anti-tumor agent in animal models and cell culture systems. Crocetin affects the growth of cancer cells by inhibiting nucleic acid synthesis, enhancing anti-oxidative system, inducing apoptosis and hindering growth factor signaling pathways. This review discusses the studies on cancer preventive potential of crocetin and its future use as an anticancer agent.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Carotenoids/therapeutic use , Neoplasms/drug therapy , Animals , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carotenoids/pharmacology , Crocus , Humans , Neoplasms/prevention & control , Vitamin A/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...