Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Publication year range
1.
Pharm Res ; 37(3): 44, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31993760

ABSTRACT

PURPOSE: This prospective study aimed to evaluate the effects of genetic polymorphisms in sulindac-related metabolizing enzyme genes including FMO3 and AOX1 on the population pharmacokinetics of sulindac in 58 pregnant women with preterm labor. METHODS: Plasma samples were collected at 1.5, 4, and 10 h after first oral administration of sulindac. Plasma concentrations of sulindac and its active metabolite (sulindac sulfide) were determined, and pharmacokinetic analysis was performed with NONMEM 7.3. RESULTS: The mean maternal and gestational ages at the time of dosing were 32.5 ± 4.4 (range, 20-41) years and 27.4 ± 4.4 (range, 16.4-33.4) weeks, respectively. In the population pharmacokinetic analysis, one depot compartment model of sulindac with absorption lag time best described the data. The metabolism of sulindac and sulindac sulfide was described using Michaelis-Menten kinetics. In stepwise modeling, gestational age impacted volume of distribution (Vc), and FMO3 rs2266782 was shown by the Michaelis constant to affect conversion of sulindac sulfide to sulindac (KM32); these were retained in the final model. CONCLUSIONS: Genetic polymorphisms of FMO3 and AOX1 could affect the pharmacokinetics of sulindac in women who undergo preterm labor. The results of this study could help clinicians develop individualized treatment plans for administering sulindac.


Subject(s)
Aldehyde Oxidase/genetics , Anti-Inflammatory Agents/pharmacokinetics , Obstetric Labor, Premature/metabolism , Oxygenases/genetics , Polymorphism, Genetic/physiology , Sulindac/pharmacokinetics , Adult , Aldehyde Oxidase/metabolism , Female , Genotype , Gestational Age , Humans , Models, Biological , Oxygenases/metabolism , Pregnancy , Prospective Studies , Signal Transduction , Sulindac/analogs & derivatives , Sulindac/metabolism
2.
Drug Metab Dispos ; 45(5): 497-500, 2017 05.
Article in English | MEDLINE | ID: mdl-28258069

ABSTRACT

Common marmosets (Callithrix jacchus) are potentially primate models for preclinical drug metabolism studies because there are similarities in the molecular characteristics of cytochrome P450 enzymes between this species and humans. However, characterization of non-cytochrome P450 enzymes has not been clarified in marmosets. Here, we report characterization of flavin-containing monooxygenases FMO1-FMO5 identified in marmoset tissues. Marmoset FMO forms shared high amino acid sequence identities (93%-95%) and phylogenetic closeness with human homologous FMO forms. FMO1 and FMO3 mRNA were abundantly expressed in the liver and kidneys among five marmoset tissues examined, where FMO3 protein was detected by immunoblotting. FMO inhibition assays using preheated tissue microsomes indicated that benzydamine N-oxygenation and sulindac sulfide S-oxygenation in the marmoset liver was mainly catalyzed by FMO3, the major hepatic FMO. Marmoset FMO3 protein heterologously expressed in Escherichia coli effectively catalyzed benzydamine N-oxygenation and sulindac sulfide S-oxygenation comparable to marmoset liver microsomes. These results indicate that the FMO3 enzyme expressed in marmoset livers mainly metabolizes benzydamine and sulindac sulfide (typical human FMO substrates), suggesting its importance for FMO-dependent drug metabolism in marmosets.


Subject(s)
Benzydamine/pharmacokinetics , Callithrix , Liver/enzymology , Oxygenases/metabolism , Sulindac/analogs & derivatives , Amino Acid Sequence , Animals , Callithrix/genetics , Callithrix/metabolism , Drug Evaluation, Preclinical/methods , Escherichia coli/genetics , Female , Hot Temperature , Humans , Male , Microsomes, Liver/enzymology , Organ Specificity , Oxygenases/genetics , Sequence Homology, Amino Acid , Species Specificity , Sulindac/pharmacokinetics
3.
Pharm Res ; 32(5): 1663-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25392229

ABSTRACT

PURPOSE: The purpose of the study was to evaluate the metabolism, pharmacokinetics and efficacy of phospho-NSAIDs in Ces1c-knockout mice. METHODS: Hydrolysis of phospho-NSAIDs by Ces1c was investigated using Ces1c-overexpressing cells. The rate of phospho-NSAID hydrolysis was compared between wild-type, Ces1c+/- and Ces1c-/- mouse plasma in vitro, and the effect of plasma Ces1c on the cytotoxicity of phospho-NSAIDs was evaluated. Pharmacokinetics of phospho-sulindac was examined in wild-type and Ces1c-/- mice. The impact of Ces1c on the efficacy of phospho-sulindac was investigated using lung and pancreatic cancer models in vivo. RESULTS: Phospho-NSAIDs were extensively hydrolyzed in Ces1c-overexpressing cells. Phospho-NSAID hydrolysis in wild-type mouse plasma was 6-530-fold higher than that in the plasma of Ces1c-/- mice. Ces1c-expressing wild-type mouse serum attenuated the in vitro cytotoxicity of phospho-NSAIDs towards cancer cells. Pharmacokinetic studies of phospho-sulindac using wild-type and Ces1c-/- mice demonstrated 2-fold less inactivation of phospho-sulindac in the latter. Phospho-sulindac was 2-fold more efficacious in inhibiting the growth of lung and pancreatic carcinoma in Ces1c -/- mice, as compared to wild-type mice. CONCLUSIONS: Our results indicate that intact phospho-NSAIDs are the pharmacologically active entities and phospho-NSAIDs are expected to be more efficacious in humans than in rodents due to their differential expression of carboxylesterases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/therapeutic use , Aspirin/analogs & derivatives , Carboxylic Ester Hydrolases/genetics , Carcinoma, Lewis Lung/drug therapy , Ibuprofen/analogs & derivatives , Organophosphates/therapeutic use , Organophosphorus Compounds/therapeutic use , Sulindac/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Aspirin/metabolism , Aspirin/pharmacokinetics , Aspirin/therapeutic use , Carboxylic Ester Hydrolases/blood , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Ibuprofen/metabolism , Ibuprofen/pharmacokinetics , Ibuprofen/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organophosphates/metabolism , Organophosphates/pharmacokinetics , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacokinetics , Sulindac/metabolism , Sulindac/pharmacokinetics , Sulindac/therapeutic use
4.
Drug Metab Dispos ; 42(1): 40-3, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24173915

ABSTRACT

This study aimed to investigate the effects of polymorphisms of the flavin-containing mono-oxygenase 3 (FMO3) and flavin-containing mono-oxygenase 6 (FMO6) genes on the pharmacokinetics of sulindac sulfide, the active metabolite of sulindac, in patients with preterm labor. Ten single-nucleotide polymorphisms (SNPs) were genotyped, and plasma sulindac sulfide concentrations were measured at 0, 1.5, 4, and 10 hours after drug administration. The area under the curve from time 0 to the last sampling time point (AUC(last)) for sulindac sulfide was obtained. The AUC(last) of sulindac sulfide was significantly higher in patients with variant-type homozygotes of FMO3 (rs909530) than those with ancestral alleles or heterozygotes. FMO3 (rs2266780) was in complete linkage disequilibrium with FMO6 (rs7885012), and there was marginal significance between the genotypes (P = 0.049). From multiple linear regression models, FMO3 (rs909530) was found to have significant influence on the AUClast of sulindac sulfide after adjusting for gestational age, weight, and all studied SNPs. The predictive contribution of rs909530 to the variability of sulindac sulfide AUC(last) was 27.0%. In conclusion, the results of this study could help clinicians predict the efficacies and side effects of sulindac in the development of individualized treatment of patients with preterm labor.


Subject(s)
Obstetric Labor, Premature/genetics , Oxygenases/genetics , Polymorphism, Single Nucleotide/genetics , Sulindac/analogs & derivatives , Adult , Area Under Curve , Female , Genotype , Gestational Age , Homozygote , Humans , Linkage Disequilibrium/genetics , Pregnancy , Sulindac/blood , Sulindac/pharmacokinetics , Young Adult
5.
Pharm Res ; 30(6): 1471-82, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23483440

ABSTRACT

PURPOSE: Formulate phospho-sulindac (P-S, OXT-328) in a Pluronic hydrogel to be used as a topical anti-inflammatory agent and study its efficacy, safety and pharmacokinetics in an arthritis model. METHODS: LEW/crlBR rats with Freund's adjuvant-induced arthritis were treated with P-S formulated in Pluronic hydrogel (PSH). We determined the clinical manifestations of arthritis including the locomotor activity of the rats; evaluated joints for inflammation, bone resorption, cartilage damage, COX-2 expression and NF-κB activation; assayed plasma IL-6 and IL-10 levels; and studied the pharmacokinetics of P-S in rats after topical or oral administration. RESULTS: PSH applied at the onset of arthritis or when arthritis was fully developed, suppressed it by 56-82%, improved the locomotor activity of the rats 2.1-4.4 fold, suppressed synovial inflammation, bone resorption, cartilage damage, NF-κB activation and COX-2 expression but not plasma IL-6 and IL-10 levels. There were no side effects. PSH produced rapidly high local levels of P-S with <14% of P-S reaching the circulation, while orally administered P-S was rapidly metabolized generating much lower joint levels of P-S. CONCLUSIONS: Topical application of PSH is efficacious and safe in the treatment of Freund's adjuvant-induced arthritis; has a favorable pharmacokinetic profile; and likely acts by suppressing key pro-inflammatory signaling pathways.


Subject(s)
Arthritis, Experimental/drug therapy , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacokinetics , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/pharmacokinetics , Sulindac/analogs & derivatives , Sulindac/administration & dosage , Sulindac/pharmacokinetics , Administration, Oral , Administration, Topical , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Arthritis, Experimental/blood , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Bone Resorption/blood , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cartilage/drug effects , Cartilage/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Female , Inflammation/blood , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-10/blood , Interleukin-10/metabolism , Interleukin-6/blood , Interleukin-6/metabolism , Joints/drug effects , Joints/metabolism , Motor Activity/drug effects , NF-kappa B/metabolism , Rats , Rats, Inbred Lew , Synovial Membrane/drug effects , Synovial Membrane/metabolism
6.
Cancer ; 118(23): 5848-56, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22605570

ABSTRACT

BACKGROUND: Reduced melanoma risk has been reported with regular use of nonsteroidal anti-inflammatory drugs (NSAIDs). However, the ability of NSAIDs to reach melanocytes in vivo and modulate key biomarkers in preneoplastic lesions such as atypical nevi has not been evaluated. METHODS: This randomized, double-blind, placebo-controlled trial of sulindac was conducted in individuals with atypical nevi (AN) to determine bioavailability of sulindac and metabolites in nevi and effect on apoptosis and vascular endothelial growth factor A (VEGFA) expression in AN. Fifty subjects with AN ≥ 4 mm in size and 1 benign nevus (BN) were randomized to sulindac (150 mg twice a day) or placebo for 8 weeks. Two AN were randomized for baseline excision, and 2 AN and BN were excised after intervention. RESULTS: Postintervention sulindac, sulindac sulfone, and sulindac sulfide concentrations were 0.31 ± 0.36, 1.56 ± 1.35, and 2.25 ± 2.24 µg/mL in plasma, and 0.51 ± 1.05, 1.38 ± 2.86, and 0.12 ± 0.12 µg/g in BN, respectively. Sulindac intervention did not significantly change VEGFA expression but did increase expression of the apoptotic marker cleaved caspase-3 in AN (increase of 3 ± 33 in sulindac vs decrease of 25 ± 45 in the placebo arm, P = .0056), although significance was attenuated (P = .1103) after adjusting for baseline expression. CONCLUSIONS: Eight weeks of sulindac intervention resulted in high concentrations of sulindac sulfone, a proapoptotic metabolite, in BN but did not effectively modulate VEGFA and cleaved caspase-3 expression. Study limitations included limited exposure time to sulindac and the need to optimize a panel of biomarkers for NSAID intervention studies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Melanoma/prevention & control , Nevus/drug therapy , Skin Neoplasms/prevention & control , Sulindac/therapeutic use , Adult , Caspase 3/metabolism , Double-Blind Method , Female , Humans , Male , Medication Adherence , Middle Aged , Sulindac/pharmacokinetics , Vascular Endothelial Growth Factor A/analysis
7.
Pharm Res ; 29(11): 3090-101, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22723123

ABSTRACT

PURPOSE: To evaluate the antitumor efficacy of solid lipid nanoparticle-encapsulated phospho-sulindac (SLN-PS) in human lung cancer. METHODS: PS was incorporated into SLNs using the emulsion evaporation technique. We determined the antitumor activity of SLN-PS in cultured lung cancer cells. The performance of SLN-PS was further evaluated by pharmacokinetic studies in mice and in a model of human lung cancer xenografts in nude mice. RESULTS: SLN-PS was >4-fold more potent than PS in inhibiting the growth of A549 and H510 cells in vitro. SLN-PS enhanced cellular uptake and facilitated PS accumulation in mitochondria, leading to oxidative stress and apoptosis via the mitochondrial-apoptosis pathway. SLN-PS was highly effective in suppressing the growth of A549 xenografts (78% inhibition compared to control, p < 0.01); while PS had no significant effect. Formulation of PS in SLNs resulted in improved pharmacokinetics in mice and an enhanced (≈ 14-fold) accumulation of PS and its metabolites in A549 xenografts. Finally, SLN-PS enhanced urinary F2-isoprostane uniquely in mice bearing A549 xenografts compared to untreated controls, suggesting that SLN-PS specifically induced oxidative stress in tumors. CONCLUSIONS: Our results show that SLN-PS is efficacious in suppressing the growth of lung cancer and merits further evaluation.


Subject(s)
Antineoplastic Agents/pharmacology , Lipids/administration & dosage , Lung Neoplasms/drug therapy , Mitochondria/drug effects , Nanoparticles/administration & dosage , Organophosphorus Compounds/pharmacology , Sulindac/analogs & derivatives , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Chemistry, Pharmaceutical , Female , Humans , Lipids/chemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Mitochondria/metabolism , Nanoparticles/chemistry , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Sulindac/administration & dosage , Sulindac/chemistry , Sulindac/pharmacokinetics , Sulindac/pharmacology , Superoxides/metabolism , Tissue Distribution , Transplantation, Heterologous , Xenograft Model Antitumor Assays
8.
Gut ; 60(3): 350-60, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20980345

ABSTRACT

BACKGROUND AND AIMS: The non-steroidal anti-inflammatory drug sulindac is an effective chemopreventive agent in sporadic colorectal cancer but its potential benefit in mismatch repair deficient cancers remains to be defined. We wanted to determine whether genetic defects that are relevant for colorectal cancer, such as Msh2 or p53 deficiency, would influence the efficiency of sulindac chemoprevention or increase the side effects. METHODS: Msh2 or p53 deficient and wild-type mice received feed containing 160-320 ppm sulindac for up to 25 weeks with or without a concurrent treatment with the carcinogen azoxymethane. Colon tissue was analysed by histopathology and molecular biology methods. RESULTS: We show that sulindac prevented azoxymethane-induced distal colon tumours in all mice. In the proximal colon, however, sulindac induced new inflammatory lesions on the mucosal folds, which further developed into adenocarcinoma in up to 18-25% of the p53 or Msh2 deficient mice but rarely in wild-type mice. This region in the proximal colon was characterised by a distinct profile of pro- and anti-inflammatory factors, which were modulated by the sulindac diet, including upregulation of hypoxia inducible factor 1α and macrophage inflammatory protein 2. CONCLUSIONS: These data show that the sulindac diet promotes carcinogenesis in the mouse proximal colon possibly through chronic inflammation. Sulindac has both beneficial and harmful effects in vivo, which are associated with different microenvironments within the colon of experimental mice. Deficiency for the Msh2 or p53 tumour suppressor genes increases the harmful side effects of long-term sulindac treatment in the mouse colon.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/therapeutic use , Colonic Neoplasms/chemically induced , Colonic Neoplasms/prevention & control , Sulindac/therapeutic use , Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anticarcinogenic Agents/adverse effects , Anticarcinogenic Agents/pharmacokinetics , Apoptosis/drug effects , Azoxymethane , Carcinogens , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , MutS Homolog 2 Protein/deficiency , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Sulindac/adverse effects , Sulindac/pharmacokinetics , Tumor Suppressor Protein p53/deficiency
9.
Drug Metab Dispos ; 39(6): 1014-21, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21383205

ABSTRACT

Sulindac is a nonsteroidal, anti-inflammatory drug (NSAID) that has also been studied for its anticancer activity. Recent studies suggest that sulindac and its metabolites act by sensitizing cancer cells to oxidizing agents and drugs that affect mitochondrial function, resulting in the production of reactive oxygen species and death by apoptosis. In contrast, normal cells are not killed under these conditions and, in some instances, are protected against oxidative stress. Sulindac has a methyl sulfoxide moiety with a chiral center and was used in all of the previous studies as a mixture of the R- and S-epimers. Because epimers of a compound can have very different chemical and biological properties, we have separated the R- and S-epimers of sulindac, studied their individual metabolism, and performed preliminary experiments on their effect on normal and lung cancer cells exposed to oxidative stress. Previous results had indicated that the reduction of (S)-sulindac to sulindac sulfide, the active NSAID, was catalyzed by methionine sulfoxide reductase (Msr) A. In the present study, we purified an enzyme that reduces (R)-sulindac and resembles MsrB in its substrate specificity. The oxidation of both epimers to sulindac sulfone is catalyzed primarily by the microsomal cytochrome P450 (P450) system, and the individual enzymes responsible have been identified. (S)-Sulindac increases the activity of the P450 system better than (R)-sulindac, but both epimers increase primarily the enzymes that oxidize (R)-sulindac. Both epimers can protect normal lung cells against oxidative damage and enhance the killing of lung cancer cells exposed to oxidative stress.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Methionine Sulfoxide Reductases/metabolism , Sulindac/metabolism , Sulindac/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Blotting, Western , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Hep G2 Cells , Humans , Methionine Sulfoxide Reductases/chemistry , Methionine Sulfoxide Reductases/genetics , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Molecular Structure , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Stereoisomerism , Sulindac/chemistry , Sulindac/pharmacokinetics
10.
J Ocul Pharmacol Ther ; 37(6): 321-330, 2021.
Article in English | MEDLINE | ID: mdl-34152861

ABSTRACT

Purpose: Inflammation of the ocular surface is central to dry eye disease (DED). The anti-inflammatory agent phospho-sulindac (PS) at a high dose was efficacious against DED in a rabbit model. We assessed the dose, formulation and structure dependence of PS's effect. Methods: In rabbits with concanavalin A-induced DED we evaluated a range of PS concentrations (0.05%-1.6%) and dosing frequencies, assessed the duration of its effect with PS in 2 solution formulations and one emulsion formulation, and compared the efficacy of PS to that of sulindac, and of the structurally similar phospho-ibuprofen amide. We determined tear breakup time (TBUT) (tear stability), Schirmer's tear test (tear production), and by esthesiometry corneal sensitivity (symptoms). We also determined the biodistribution in the eye of topically applied PS. Results: PS in a solution formulation, given as eye drops q.i.d. was efficacious starting at a dose of 0.1%. The effect was apparent after 2 days of treatment and lasted at least 8 days after the last dose. Both signs (evidenced by TBUT and Schirmer's test) and symptoms (measured by corneal sensitivity) improved significantly. The best formulation was the solution formulation; a cyclodextrin-based formulation was also successful but the emulsion formulation was not. PS and its metabolites were essentially restricted to the anterior chamber of the eye. Sulindac and phospho-ibuprofen amide had no efficacy on DED. Conclusions: PS is efficacious against DED. Its effect, encompassing signs, and symptoms, are dose, formulation, and structure dependent. PS has therapeutic promise and merits further development.


Subject(s)
Drug Compounding , Dry Eye Syndromes/drug therapy , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Sulindac/analogs & derivatives , Administration, Topical , Animals , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Male , Organophosphorus Compounds/pharmacokinetics , Rabbits , Sulindac/administration & dosage , Sulindac/chemistry , Sulindac/pharmacokinetics , Tissue Distribution
11.
Int J Pharm ; 557: 273-279, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30597269

ABSTRACT

We studied the pharmacokinetics, biodistribution and metabolism of phospho-sulindac (PS), a novel agent efficacious in the treatment of dry eye, formulated in nanoparticles (PS-NPs) following its topical administration to the eye of New Zealand White rabbits. The nanoparticles were spherical with effective diameter = 108.9 ±â€¯41.7 nm, zeta potential = -21.70 ±â€¯3.78 mV, drug loading = 7%, and entrapment efficiency = 46.4%. Of the total PS delivered topically to the eye, >95% was retained in the anterior segment, predominantly in the cornea (Cmax = 101.3 µM; Tmax = 1 h; T1/2 = 2.6 h; area AUC0-16h = 164.4 µM·h) and conjunctiva (Cmax = 89.4 µM; Tmax = 0.25 h; T1/2 = 3.1 h; AUC0-16h = 63.5 µM·h), the tissues most affected by dry eye disease. No PS or its metabolites were detected in the systemic circulation. PS was metabolized to PS sulfide and PS sulfone; all three molecules were hydrolyzed to sulindac, which was converted to sulindac sulfide and sulindac sulfone. A solution formulation of PS provided lower PS levels in ocular tissues but higher levels of PS metabolites, compared to PS-NPs. Therefore, NPs represent an effective formulation for the topical ocular administration of PS for anterior segment diseases, such as dry eye disease.


Subject(s)
Drug Delivery Systems , Eye/metabolism , Nanoparticles/administration & dosage , Organophosphorus Compounds/administration & dosage , Sulindac/analogs & derivatives , Administration, Intravenous , Administration, Topical , Animals , Male , Nanoparticles/chemistry , Organophosphorus Compounds/blood , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Rabbits , Sulindac/administration & dosage , Sulindac/blood , Sulindac/chemistry , Sulindac/pharmacokinetics , Tissue Distribution
12.
Cancer Epidemiol Biomarkers Prev ; 17(3): 674-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18349286

ABSTRACT

The cyclooxygenase (COX)-2 enzyme appears to be an important target for cancer chemoprevention. Given the recent emergence of potentially serious cardiovascular toxicity associated with selective COX-2 inhibitors, nonsteroidal antiinflammatory drugs (NSAIDs), which inhibit both COX-1 and COX-2, have received renewed attention as candidate chemoprevention agents. Sulindac has shown consistent chemopreventive potential in preclinical studies as well as in a limited number of clinical trials reported to date. For the current pharmacokinetic study, sulindac capsules were prepared to facilitate ample agent supplies for future intervention studies. Encapsulation of the parent compound (sulindac sulfoxide) can be readily accomplished, but the effects of alternate formulations on bioavailability have not been rigorously examined. In the present single-dose, two-period crossover trial, we conducted pharmacokinetic analyses of sulindac in capsule (test) versus tablet (reference) formulations. Overall, bioavailability appeared to be higher for the capsule compared with the tablet formulation based on test-to-reference pharmacokinetic variable ratios for the parent compound alone. However, additional analyses based on the sulfide and sulfone metabolites of sulindac with the same pharmacokinetic variables indicated similar chemopreventive exposures between the capsule and tablet formulations. These data support the use of sulindac capsules, which can be readily prepared with matching placebos, in future blinded chemoprevention trials.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Sulindac/pharmacokinetics , Analysis of Variance , Antineoplastic Agents/administration & dosage , Area Under Curve , Biological Availability , Capsules , Chromatography, High Pressure Liquid , Cross-Over Studies , Female , Humans , Male , Middle Aged , Sulindac/administration & dosage , Tablets
13.
Cancer Chemother Pharmacol ; 59(1): 79-87, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16642371

ABSTRACT

PURPOSE: Multi-drug resistance mediated by ATP-binding cassette trans-membrane protein pumps is an important cause of cancer treatment failure. Sulindac has been shown to be a competitive substrate for the clinically important resistance protein, multi-drug resistance protein-1 (MRP-1), and thus might enhance the anti-cancer activity of substrate chemotherapeutic agents, e.g. anthracyclines. METHODS: We conducted a dose-escalating, single arm, prospective, open label, non-randomised phase I trial of epirubicin (75 mg/m(2)) in combination with escalating oral doses of sulindac (0-800 mg) in patients with advanced cancer to identify an appropriate dose of sulindac to use in future resistance studies. Anthracycline and sulindac pharmacokinetics were studied in cycles 1 and 3. RESULTS: Seventeen patients (8 breast, 3 lung, 2 bowel, 1 melanoma, 1 renal, 1 ovarian and 1 of unknown primary origin, 16/17 having had prior chemotherapy) were enrolled. Eight patients received a full six cycles of treatment; 14 patients received three or more cycles. Dose-limiting toxicity was observed in two patients at 800 mg sulindac (1 renal impairment, 1 fatal haemoptysis in a patient with advanced lung cancer), and sulindac 600 mg was deemed to be the maximum tolerated dose. Sulindac had no effect on epirubicin pharmacokinetics. Among 15 patients with evaluable tumour, two partial responses were seen (malignant melanoma and breast cancer). Four others had prolonged stable disease. CONCLUSION: Epirubicin 75 mg/m(2) and sulindac 600 mg are the recommended doses for phase II studies for these agents in combination.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Epirubicin/therapeutic use , Neoplasms/drug therapy , Sulindac/pharmacokinetics , Sulindac/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Aged , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antibiotics, Antineoplastic/adverse effects , Chemotherapy, Adjuvant , Creatinine/blood , Dose-Response Relationship, Drug , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Epirubicin/adverse effects , Female , Humans , Immunohistochemistry , Male , Middle Aged , Myocardium/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Platelet Count , Prospective Studies , Sulindac/adverse effects , Troponin/metabolism
14.
Biomed Res Int ; 2017: 4189678, 2017.
Article in English | MEDLINE | ID: mdl-28331852

ABSTRACT

Sulindac is a nonsteroidal anti-inflammatory drug, which is clinically used for the ailments of various inflammations. This study investigated the allele frequencies of FMO3 E158K and E308G and evaluated the influences of these two genetic polymorphisms on the pharmacokinetics of sulindac and its metabolites in Chinese healthy male volunteers. Eight FMO3 wild-type (FMO3 HHDD) subjects and seven FMO3 homozygotes E158K and E308G mutant (FMO3 hhdd) subjects were recruited from 247 healthy male volunteers genotyped by PCR-RFLP method. The plasma concentrations of sulindac, sulindac sulfide, and sulindac sulfone were determined by UPLC, while the pharmacokinetic parameters of the two different FMO3 genotypes were compared with each other. The frequencies of FMO3 E158K and E308G were 20.3% and 20.1%, respectively, which were in line with Hardy-Weinberg equilibrium (D' = 0.977, r2 = 0.944). The mean values of Cmax, AUC0-24, and AUC0-∞ of sulindac were significantly higher in FMO3 hhdd group than those of FMO3 HHDD group (P < 0.05), while the pharmacokinetic parameters except Tmax of sulindac sulfide and sulindac sulfone showed no statistical difference between the two groups. The two FMO3 mutants were in close linkage disequilibrium and might play an important role in the pharmacokinetics of sulindac in Chinese healthy male volunteers.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Inflammation/drug therapy , Oxygenases/genetics , Sulindac/administration & dosage , Adult , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Gene Frequency , Genotype , Healthy Volunteers , Humans , Inflammation/genetics , Linkage Disequilibrium , Male , Polymorphism, Restriction Fragment Length , Sulindac/analogs & derivatives , Sulindac/pharmacokinetics
15.
J Chromatogr A ; 1119(1-2): 176-82, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16530777

ABSTRACT

Field-amplified sample stacking with electroosmotic flow (EOF) suppressant in capillary electrophoresis was used to determine the concentration of sulindac (SU) and its two active metabolites, sulindac sulfide (SI) and sulindac sulfone (SO), in human plasma. After acidification, the analytes were extracted from the plasma with dichloromethane. Before sample loading, a water plug (0.5 psi, 3 s) was injected to contain sample anions and to permit field-amplified stacking. Electrokinetic injection at a reversed voltage (-6 kV, 99.9 s) was then used to introduce anions. Separation was performed using phosphate buffer (80 mM, pH 6.0) containing 2,6-di-O-methyl-beta-cyclodextrin (0.75 mM), and poly(ethylene oxide) (0.01%) as an EOF suppressant. The separation was performed at -30 kV and 200 nm. During method validation, calibration plots were linear (r > 0.994) over a range of 0.3-30.0 microM for SU and SO, and 0.5-30.0 microM for SI. During intra- and inter-day analysis, relative standard deviations (RSD) and relative errors (RE) were all less than 16%. The limits of detection were 0.1 microM for SU and SO, and 0.3 microM for SI (S/N = 4, sampling 99.9s at -6 kV). This method was feasible for determining SU and its metabolites in plasma. One female volunteer (27 years, 42 kg) was orally administered one SU tablet (Clinoril, 20 0 mg/tab), and blood samples were drawn at regular intervals over an 8h period. After pretreatment and analysis, the plasma levels of SU, SI and SO were monitored. The pharmacokinetic profile of SU was also investigated.


Subject(s)
Electrophoresis, Capillary/methods , Sulindac/blood , Adult , Female , Humans , Hydrogen-Ion Concentration , Osmosis , Polyethylene Glycols , Sulindac/analogs & derivatives , Sulindac/pharmacokinetics , beta-Cyclodextrins
16.
Chem Biol Interact ; 164(1-2): 68-75, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17027946

ABSTRACT

In cancer chemopreventive studies, test agents are typically administered via diet, while the preclinical safety studies normally employ oral gavage dosing. Correspondence in pharmacokinetic and pharmacodynamic profiles between the two dosing approaches cannot be assumed a priori. Sulindac, a non-steroidal anti-inflammatory agent with potential chemopreventive activity, was used to assess effects of the two oral dosing paradigms on its pharmacokinetics and pharmacodynamics. Time-dependent concentrations of sulindac and its sulfone metabolite were determined in plasma and potential target organ, mammary gland. Prostaglandin E(2) was used as a pharmacodynamic biomarker and measured in mammary gland. An inverse linear relationship was detected between pharmacodynamic and pharmacokinetic markers, area under the curve for prostaglandin E(2) levels and sulindac sulfone concentrations, respectively, in the mammary tissue. Marked differences in pharmacokinetics and pharmacodynamics were observed after administration of sulindac by the two oral dosing paradigms. In general, oral gavage resulted in higher peak and lower trough concentrations of sulindac in plasma and mammary tissue, higher area under concentration-time curve in plasma and mammary tissue, and greater effect on prostaglandin E(2) levels than the corresponding diet dosing. This study illustrates potential pitfalls and limitations in trying to generalize based on data obtained with different oral dosing schemes and their extrapolation to potential efficacy and health risks in humans.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Drug Administration Schedule/veterinary , Mammary Glands, Animal/metabolism , Sulindac/pharmacokinetics , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Antineoplastic Agents/administration & dosage , Biomarkers/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Female , Rats , Rats, Sprague-Dawley , Risk Factors , Sulindac/administration & dosage , Sulindac/analogs & derivatives , Time Factors
17.
Cancer Res ; 57(14): 2909-15, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9230200

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs), such as sulindac, have cancer chemopreventive properties by a mechanism that has been suggested to involve cyclooxygenase inhibition and reduction of prostaglandin (PGE2) levels in the target tissue. To test this hypothesis, we studied the effect of dietary sulindac sulfone (500-2000 ppm), a metabolite of sulindac reported to lack cyclooxygenase inhibitory activity, on tumor formation and PGE2 levels in the azoxymethane model of colon carcinogenesis. Rats treated with sulindac at 400 ppm and piroxicam at 150 ppm were used as positive controls. Rats received two s.c. injections of azoxymethane (15 mg/kg) for 2 weeks and were fed either experimental or control diets until necropsy. After 31 weeks of sulfone treatment, a dose-related increase in sulfone levels in both serum and cecal contents was measured; there was no evidence of metabolic conversion to sulindac or other metabolites. Rats treated with sulfone at 1000 and 2000 ppm, sulindac, and piroxicam had significantly fewer colonic adenomas and carcinomas compared with rats fed control diet as measured by tumor incidence, multiplicity, and tumor burden. Sulfone-treated rats also showed a dose-response relationship for inhibiting all tumor parameters. Colons from rats treated with sulindac or piroxicam contained PGE2 levels that ranged from approximately 16-49% of control levels. PGE2 levels in rats treated with sulfone up to 2000 ppm ranged from 78-118% of control levels. Moreover, the effects of sulindac sulfone on various enzymes responsible for regulating prostaglandin levels were evaluated. No significant inhibitory effects were observed for cyclooxygenase, lipoxygenase, or phospholipase A2. These results suggest that reduction of prostaglandin levels in the target tissue may not be necessary for the chemopreventive properties of sulindac.


Subject(s)
Anticarcinogenic Agents/pharmacology , Azoxymethane/toxicity , Carcinogens/toxicity , Colonic Neoplasms/prevention & control , Dinoprostone/analysis , Sulindac/analogs & derivatives , Animals , Colonic Neoplasms/chemically induced , Male , Rats , Rats, Inbred F344 , Sulindac/pharmacokinetics , Sulindac/pharmacology
18.
Cancer Res ; 61(4): 1733-40, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245490

ABSTRACT

Mutations in the human adenomatous polyposis (APC) gene are causative for familial adenomatous polyposis (FAP), a rare condition in which numerous colonic polyps arise during puberty and, if left untreated, lead to colon cancer. The APC gene is a tumor suppressor that has been termed the "gatekeeper gene" for colon cancer. In addition to the 100% mutation rate in FAP patients, the APC gene is mutated in >80% of sporadic colon and intestinal cancers. The Apc gene in mice has been mutated either by chemical carcinogenesis, resulting in the Min mouse Apcdelta850, or by heterologous recombination, resulting in the Apcdelta716 or Apedelta1368 mice (M. Oshima et al., Proc. Natl. Acad. Sci. USA, 92: 4482-4486, 1995). Although homozygote Apc-/- mice are embryonically lethal, the heterozygotes are viable but develop numerous intestinal polyps with loss of Apc heterozygosity within the polyps (M. Oshima et al., Proc. Natl. Acad. Sci. USA, 92: 4482-4486, 1995). The proinflammatory, prooncogenic protein cyclooxygenase (COX)-2 has been shown to be markedly induced in the Apcdelta716 polyps at an early stage of polyp development (M. Oshima et al., Cell, 87: 803-809, 1996). We demonstrate here that treatment with the specific COX-2 inhibitor rofecoxib results in a dose-dependent reduction in the number and size of intestinal and colonic polyps in the Apcdelta716 mouse. The plasma concentration of rofecoxib that resulted in a 55% inhibition of polyp number and an 80% inhibition of polyps > 1 mm in size is comparable with the human clinical steady-state concentration of 25 mg rofecoxib (Vioxx) taken once daily (A. Porras et al., Clin. Pharm. Ther., 67: 137, 2000). Polyps from both untreated and rofecoxib- or sulindac-treated Apcdelta716 mice expressed COX-1 and -2, whereas normal epithelium from all mice expressed COX-1 but minimal amounts of COX-2. Polyps from either rofecoxib- or sulindac-treated mice had lower rates of DNA replication, expressed less proangiogenic vascular endothelial-derived growth factor and more membrane-bound beta-catenin, but showed unchanged nuclear localization of this transcription factor. This study showing the inhibition of polyposis in the Apcdelta716 mouse suggests that the specific COX-2 inhibitor rofecoxib (Vioxx) has potential as a chemopreventive agent in human intestinal and colon cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Genes, APC/genetics , Intestinal Neoplasms/prevention & control , Intestinal Polyps/prevention & control , Isoenzymes/antagonists & inhibitors , Lactones/pharmacology , Trans-Activators , Animals , Anticarcinogenic Agents/pharmacokinetics , Cell Nucleus/metabolism , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacokinetics , Cytoskeletal Proteins/metabolism , DNA Replication/drug effects , Dose-Response Relationship, Drug , Female , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Intestinal Polyps/enzymology , Intestinal Polyps/genetics , Isoenzymes/biosynthesis , Lactones/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/biosynthesis , Sulfones , Sulindac/analogs & derivatives , Sulindac/pharmacokinetics , Sulindac/pharmacology , beta Catenin
19.
Cancer Res ; 59(14): 3387-91, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10416599

ABSTRACT

Epidemiological and model studies with laboratory animals have provided evidence that nonsteroidal anti-inflammatory drugs reduce the risk of colon cancer. Sulindac, a nonsteroidal anti-inflammatory drug, has been shown to inhibit azoxymethane (AOM)-induced colon carcinogenesis in rats when administered continuously before, during, and after carcinogen treatment (initiation and postinitiation periods) or when given continuously beginning 14 weeks after carcinogen administration (promotion/ progression stage). The present study was designed to investigate the chemopreventive efficacy of sulindac sulfone (exisulind), the sulfone metabolite of sulindac, when administered during the promotion/progression stage of colon carcinogenesis in comparison to the effect during the initiation and postinitiation periods. We have also studied the modulating effect of exisulind on colonic tumor apoptosis. At 5 weeks of age, groups of male F344 rats were fed diets containing 0%, 0.06%, and 0.12% exisulind. At 7 weeks of age, groups of animals were injected s.c. with AOM (15 mg/kg body weight, once weekly for 2 weeks). Animals intended for the promotion/progression study and receiving 0% exisulind were switched to an experimental diet containing 0.12% exisulind at 14 weeks after the second AOM treatment. All rats remained on their respective dietary regimens until the termination of the study, 50 weeks after the second AOM injection. Colon tumors were evaluated histopathologically for tumor type. Administration of 0.06% and 0.12% exisulind during the initiation and postinitiation periods significantly inhibited the incidence and multiplicity of invasive and/or noninvasive adenocarcinomas of the colon. The inhibition of colon tumorigenesis by exisulind was associated with a significant retardation of body weight gain shortly after sulfone administration and increased apoptosis in the colon tumors. In contrast, administration of the higher dose (0.12%) of exisulind during the promotion/progression stage had only minimal effects on colon tumorigenesis and apoptosis in the colon tumors, suggesting that early administration, but not late administration, may be required for chemopreventive efficacy of this drug.


Subject(s)
Adenocarcinoma/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anticarcinogenic Agents/administration & dosage , Colonic Neoplasms/prevention & control , Sulindac/analogs & derivatives , Adenocarcinoma/chemically induced , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Apoptosis/drug effects , Azoxymethane/administration & dosage , Carcinogens/administration & dosage , Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/chemically induced , Cyclooxygenase Inhibitors/pharmacology , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Neoplasm Invasiveness , Rats , Rats, Inbred F344 , Sulindac/administration & dosage , Sulindac/pharmacokinetics , Sulindac/pharmacology , Sulindac/therapeutic use , Weight Gain/drug effects
20.
Clin Lung Cancer ; 6(6): 361-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15943897

ABSTRACT

Exisulind is a sulfone derivative of sulindac that induces apoptosis and demonstrates synergy with docetaxel in lung cancer models. This study evaluated the safety, efficacy, and pharmacokinetic interactions of exisulind and docetaxel/carboplatin in patients with metastatic non-small-cell lung cancer (NSCLC). Fifty-seven patients received 218 cycles of docetaxel (75 mg/m2) and carboplatin (area under the curve, 5.0) in combination with exisulind (125-250 mg orally twice daily). Two complete responses and 9 partial responses were observed among the 47 patients assessable for response (overall response rate, 23%). The median duration of response was 5.9 months and median survival was 9.4 months. The 1- and 2-year survival rates are 35% and 14%, respectively. The hematologic toxicities were consistent with those previously reported with docetaxel/carboplatin. The most common nonhematologic toxicities were mild to moderate fatigue, anorexia, nausea, and vomiting. The addition of exisulind to the chemotherapy regimen did not interfere with the metabolism or elimination of docetaxel and vice versa, and docetaxel did not interfere with the pharmacokinetic parameters of exisulind. This trial did not allow direct comparison of patients receiving docetaxel/carboplatin with and without exisulind, but when compared with historical data of docetaxel/carboplatin alone, the addition of exisulind does not appear to enhance antitumor activity, duration of response, or survival. Although preclinical data demonstrate increased apoptosis and prolonged survival for the combination of exisulind and docetaxel, multiple clinical trials do not support further clinical development of this combination regimen in patients with advanced NSCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Carboplatin/administration & dosage , Carboplatin/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/pathology , Docetaxel , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Sulindac/administration & dosage , Sulindac/analogs & derivatives , Sulindac/pharmacokinetics , Taxoids/administration & dosage , Taxoids/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL