Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Int J Oncol ; 43(3): 895-902, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23807084

ABSTRACT

Phospho-sulindac (PS) is a safe sulindac derivative with promising anticancer efficacy in colon cancer. We evaluated whether its combination with curcumin could enhance the efficacy in the treatment of lung cancer. Curcumin, the principal bioactive component in turmeric, has demonstrated versatile capabilities to modify the therapeutic efficacy of a wide range of anticancer agents. Here, we evaluated the effect of co-administration of curcumin on the anticancer activity of PS in a mouse xenograft model of human lung cancer. Curcumin enhanced the cellular uptake of PS in human lung and colon cancer cell lines. To assess the potential synergism between curcumin and PS in vivo, curcumin was suspended in 10% Tween-80 or formulated in micellar nanoparticles and given to mice by oral gavage prior to the administration of PS. Both formulations of curcumin significantly improved the pharmacokinetic profiles of PS, with the 10% Tween-80 suspension being much more effective than the nanoparticle formation. However, curcumin did not exhibit any significant modification of the metabolite profile of PS. Furthermore, in a mouse subcutaneous xenograft model of human lung cancer, PS (200 mg/kg) in combination with curcumin (500 mg/kg) suspended in 10% Tween-80 (51% inhibition, p<0.05) was significantly more efficacious than PS plus micelle curcumin (30%) or PS (25%) or curcumin alone (no effect). Consistent with the improved pharmacokinetics, the combination treatment group had higher levels of PS and its metabolites in the xenografts compared to PS alone. Our results show that curcumin substantially improves the pharmacokinetics of PS leading to synergistic inhibition of the growth of human lung cancer xenografts, representing a promising drug combination.


Subject(s)
Curcumin/administration & dosage , Drug Synergism , Lung Neoplasms/drug therapy , Sulindac/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Sulindac/analogs & derivatives , Sulindac/pharmacokinetics , Xenograft Model Antitumor Assays
9.
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
10.
J Clin Pharmacol ; 53(4): 403-12, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23436338

ABSTRACT

Sulindac is a prescription-based non-steroidal anti-inflammatory drug (NSAID) that continues to be actively investigated as a candidate cancer chemoprevention agent. To further current understanding of sulindac bioavailability, metabolism, and disposition, we developed a population pharmacokinetic model for the parent compound and its active metabolites, sulindac sulfide, and exisulind. This analysis was based on data from 24 healthy subjects who participated in a bioequivalence study comparing two formulations of sulindac. The complex disposition of sulindac and its metabolites was described by a seven-compartment model featuring enterohepatic recirculation and is the first reported population pharmacokinetic model for sulindac. The derived model was used to explore effects of clinical variables on sulindac pharmacokinetics and revealed that body weight, creatinine clearance, and gender were significantly correlated with pharmacokinetic parameters. Moreover, the model quantifies the relative bioavailability of the sulindac formulations and illustrates the utility of population pharmacokinetics in bioequivalence assessment. This novel population pharmacokinetic model provides new insights regarding the factors that may affect the pharmacokinetics of sulindac and the exisulind and sulindac sulfide metabolites in generally healthy subjects, which have implications for future chemoprevention trial design for this widely available agent.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Models, Biological , Sulindac/pharmacokinetics , Adult , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/blood , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Biological Availability , Capsules , Cross-Over Studies , Female , Humans , Male , Neoplasms/prevention & control , Sulindac/administration & dosage , Sulindac/analogs & derivatives , Sulindac/blood , Tablets
11.
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
12.
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
13.
Biochem Pharmacol ; 83(6): 715-22, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22222427

ABSTRACT

Hydrogen sulfide-releasing non-steroidal anti-inflammatory drugs (HS-NSAIDs) are an emerging novel class of compounds with significant anti-inflammatory properties. They consist of a traditional NSAID to which an H(2)S-releasing moiety is covalently attached. We examined the effects of four different HS-NSAIDs on the growth properties of eleven different human cancer cell lines of six different tissue origins. Human colon, breast, pancreatic, prostate, lung, and leukemia cancer cell lines were treated with HS-aspirin, -sulindac, -iburofen, -naproxen, and their traditional counterparts. HS-NSAIDs inhibited the growth of all cancer cell lines studied, with potencies of 28- to >3000-fold greater than that of their traditional counterparts. HS-aspirin (HS-ASA) was consistently the most potent. HS-NSAIDs inhibited cell proliferation, induced apoptosis, and caused G(0)/G(1) cell cycle block. Metabolism of HS-ASA by colon cells showed that the acetyl group of ASA was hydrolyzed rapidly, followed by hydrolysis of the ester bond linking the salicylate anion to the H(2)S releasing moiety, producing salicylic acid and ADT-OH from which H(2)S is released. In reconstitution studies, ASA and ADT-OH were individually less active than the intact HS-ASA towards cell growth inhibition. Additionally, the combination of these two components representing a fairly close approximation to the intact HS-ASA, was 95-fold less active than the intact HS-ASA for growth inhibition. Taken together, these results demonstrate that HS-NSAIDs have potential anti-growth activity against a wide variety of human cancer cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Aspirin/analogs & derivatives , Cell Proliferation/drug effects , Growth Inhibitors/pharmacokinetics , Hydrogen Sulfide/metabolism , Ibuprofen/analogs & derivatives , Naproxen/analogs & derivatives , Sulindac/analogs & derivatives , Thiones/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Aspirin/chemistry , Aspirin/pharmacokinetics , Cell Line, Tumor , Growth Inhibitors/chemistry , Humans , Hydrogen Sulfide/chemistry , Ibuprofen/chemistry , Ibuprofen/pharmacokinetics , Naproxen/chemistry , Naproxen/pharmacokinetics , Sulindac/chemistry , Sulindac/pharmacokinetics , Thiones/chemistry
14.
Br J Pharmacol ; 165(7): 2152-66, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21955327

ABSTRACT

BACKGROUND AND PURPOSE: Phospho-sulindac (PS; OXT-328) prevents colon cancer in mice, especially when combined with difluoromethylornithine (DFMO). Here, we explored its metabolism and pharmacokinetics. EXPERIMENTAL APPROACH: PS metabolism was studied in cultured cells, liver microsomes and cytosol, intestinal microsomes and in mice. Pharmacokinetics and biodistribution of PS were studied in mice. KEY RESULTS: PS undergoes reduction and oxidation yielding PS sulphide and PS sulphone; is hydrolysed releasing sulindac, which generates sulindac sulphide (SSide) and sulindac sulphone (SSone), all of which are glucuronidated. Liver and intestinal microsomes metabolized PS extensively but cultured cells converted only 10% of it to PS sulphide and PS sulphone. In mice, oral PS is rapidly absorbed, metabolized and distributed to the blood and other tissues. PS survives only partially intact in blood; of its three major metabolites (sulindac, SSide and SSone), sulindac has the highest C(max) and SSone the highest t(1/2) ; their AUC(0-24h) are similar. Compared with conventional sulindac, PS generated more SSone but less SSide, which may contribute to the safety of PS. In the gastroduodenal wall of mice, 71% of PS was intact; sulindac, SSide and SSone together accounted for <30% of the total. This finding may explain the lack of gastrointestinal toxicity by PS. DFMO had no effect on PS metabolism but significantly reduced drug level in mouse plasma and other tissues. CONCLUSIONS AND IMPLICATIONS: Our findings establish the metabolism of PS define its pharmacokinetics and biodistribution, describe its interactions with DFMO and largely explain its gastrointestinal safety.


Subject(s)
Eflornithine/pharmacology , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacokinetics , Sulindac/analogs & derivatives , Animals , Cell Line, Tumor , Colonic Neoplasms/prevention & control , Cytosol/metabolism , Eflornithine/administration & dosage , Female , Humans , In Vitro Techniques , Intestinal Mucosa/metabolism , Mice , Mice, Inbred BALB C , Microsomes/metabolism , Microsomes, Liver/metabolism , Organophosphorus Compounds/administration & dosage , Rats , Sulindac/administration & dosage , Sulindac/metabolism , Sulindac/pharmacokinetics , Tissue Distribution
15.
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
16.
Cancer Prev Res (Phila) ; 4(1): 150-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21205743

ABSTRACT

The antitumor effects of nonsteroidal anti-inflammatory drugs (NSAID) are assumed to be due to the inhibition of COX activity, but COX-independent mechanisms may also play an important role. NSAID-activated gene (NAG-1/GDF15) is induced by NSAIDs and has antitumorigenic activities. To determine the contribution of COX-2 inhibition and NAG-1/GDF15 expression to the prevention of colon carcinogenesis by NSAIDs, we evaluated several sulindac derivatives [des-methyl (DM)-sulindac sulfide and its prodrug DM-sulindac] that do not inhibit COX-2 activity. Sulindac sulfide and DM-sulindac induced the expression of NAG-1/GDF15 in HCT116 cells as determined by quantitative real-time PCR and Western blot. We fed APC/Min mice with 320 ppm of sulindac and doses of DM-sulindac. Only sulindac significantly inhibited tumor formation inAPC/Min mice. To determine the pharmacokinetic properties of sulindac and DM-sulindac in vivo, wild-type C57/B6 mice were fed with sulindac and DM-sulindac at 80, 160, and 320 ppm. High-performance liquid chromatography analysis revealed that the conversion of DM-sulindac to DM-sulindac sulfide (active form) was less efficient than the conversion of sulindac to sulindac sulfide (active form) in the mice. Lower levels of DM-sulindac sulfide accumulated in intestinal and colon tissues in comparison with sulindac sulfide. In addition, NAG-1/GDF15 was induced in the liver of sulindac-fed mice but not in the DM-sulindac-fed mice. Collectively, our results suggest that the tumor-inhibitory effects of sulindac in APC/Min mice may be due to, in part, NAG-1/GDF15 induction in the liver. Our study also suggests that pharmacologic properties should be carefully evaluated when developing drug candidates.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Genes, APC/physiology , Growth Differentiation Factor 15/physiology , Intestinal Polyps/drug therapy , Sulindac/administration & dosage , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Apoptosis/drug effects , Blotting, Western , Disease Models, Animal , Humans , Immunoenzyme Techniques , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulindac/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
17.
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
18.
Cancer Chemother Pharmacol ; 67(2): 431-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20445979

ABSTRACT

PURPOSE: To find the maximum tolerated dose (MTD) of OSI-461 in combination with mitoxantrone in patients with advanced solid tumors. METHODS: This was a Phase I study using cohort dose escalation of OSI-461 dosed orally twice daily in combination with mitoxantrone 12 mg/m(2) given on Day 1 of each 21-day cycle. RESULTS: OSI-461 dose was escalated to 1,000 mg po bid. One patient experienced a dose-limiting toxicity (DLT). Three patients discontinued the study due to adverse events (AE). Two patients (10%) had a partial response, and ten patients (50%) had stable disease as best response. CONCLUSION: The combination of OSI-461 and mitoxantrone was well tolerated. Dose escalation was stopped because of toxicities in a concurrent Phase I trial. The response rate seen in patients with prostate cancer was comparable to response rates seen in trials of mitoxantrone and prednisone alone, and further studies of the combination of OSI-461 and mitoxantrone were not pursued.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mitoxantrone/administration & dosage , Neoplasms/drug therapy , Sulindac/analogs & derivatives , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Breast Neoplasms/drug therapy , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Mitoxantrone/adverse effects , Mitoxantrone/pharmacokinetics , Prostatic Neoplasms/drug therapy , Sulindac/administration & dosage , Sulindac/adverse effects , Sulindac/pharmacokinetics , Testicular Neoplasms/drug therapy , Treatment Outcome , Urinary Bladder Neoplasms/drug therapy
19.
Cancer Prev Res (Phila) ; 3(1): 101-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20051377

ABSTRACT

Regular use of nonsteroidal anti-inflammatory drugs (NSAID) has been associated with reduced risk of breast cancer. Sulindac, a nonselective NSAID with both cyclooxygenase-2-dependent and -independent activities, is a candidate for breast chemoprevention. We conducted a phase Ib trial in 30 women at increased risk for breast cancer to evaluate the breast tissue distribution of sulindac at two dose levels (150 mg daily and 150 mg twice daily for 6 weeks), using nipple aspirate fluid (NAF) as a surrogate of breast tissue drug exposure. We also explored the effect of sulindac on drug-induced biomarkers in NAF. We show that sulindac and its metabolites partition to human breast as measured by NAF levels. Sulindac intervention did not decrease 13,14-dihydro-15-keto prostaglandin A(2), a stable derivative of prostaglandin E(2), in NAF, but exposure was associated with a significant trend towards higher levels of growth differentiation factor 15 in NAF in women receiving 150 mg twice daily (P = 0.038). These results are the first to show partitioning of sulindac and metabolites to human breast tissue and the first evidence for a potential dose-dependent effect of sulindac on growth differentiation factor 15 levels in NAF.


Subject(s)
Antineoplastic Agents/metabolism , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , Nipple Aspirate Fluid/metabolism , Sulindac/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Biomarkers, Tumor/analysis , Breast Neoplasms/genetics , C-Reactive Protein/analysis , C-Reactive Protein/drug effects , Carcinoma, Ductal, Breast/genetics , Chromatography, High Pressure Liquid , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Female , Genetic Predisposition to Disease , Growth Differentiation Factor 15/drug effects , Growth Differentiation Factor 15/metabolism , Humans , Nipple Aspirate Fluid/chemistry , Sulindac/administration & dosage , Sulindac/pharmacokinetics , Tissue Distribution
20.
Cancer Prev Res (Phila) ; 2(6): 572-80, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19470791

ABSTRACT

Nonsteroidal anti-inflammatory drugs such as sulindac have shown promising antineoplastic activity, although toxicity from cyclooxygenase (COX) inhibition and the suppression of prostaglandin synthesis limits their use for chemoprevention. Previous studies have concluded that the mechanism responsible for their antineoplastic activity may be COX independent. To selectively design out the COX inhibitory activity of sulindac sulfide (SS), in silico modeling studies were done that revealed the crucial role of the carboxylate moiety for COX-1 and COX-2 binding. These studies prompted the synthesis of a series of SS derivatives with carboxylate modifications that were screened for tumor cell growth and COX inhibitory activity. A SS amide (SSA) with a N,N-dimethylethyl amine substitution was found to lack COX-1 and COX-2 inhibitory activity, yet potently inhibit the growth of human colon tumor cell lines, HT-29, SW480, and HCT116 with IC(50) values of 2 to 5 micromol/L compared with 73 to 85 micromol/L for SS. The mechanism of growth inhibition involved the suppression of DNA synthesis and apoptosis induction. Oral administration of SSA was well-tolerated in mice and generated plasma levels that exceeded its in vitro IC(50) for tumor growth inhibition. In the human HT-29 colon tumor xenograft mouse model, SSA significantly inhibited tumor growth at a dosage of 250 mg/kg. Combined treatment of SSA with the chemotherapeutic drug, Camptosar, caused a more sustained suppression of tumor growth compared with Camptosar treatment alone. These results indicate that SSA has potential safety and efficacy advantages for colon cancer chemoprevention as well as utility for treating malignant disease if combined with chemotherapy.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Colonic Neoplasms/pathology , Animals , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Cell Line, Tumor/transplantation , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/drug effects , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/drug effects , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/toxicity , Drug Screening Assays, Antitumor , Humans , Irinotecan , Male , Maximum Tolerated Dose , Mice , Mice, Nude , Models, Molecular , Neoplasm Proteins/analysis , Protein Conformation , Sulindac/administration & dosage , Sulindac/analogs & derivatives , Sulindac/chemical synthesis , Sulindac/pharmacokinetics , Sulindac/pharmacology , Sulindac/therapeutic use , Sulindac/toxicity , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...