Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Pharm Res ; 41(4): 651-672, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38519817

ABSTRACT

BACKGROUND AND PURPOSE: There is concern that subvisible aggregates in biotherapeutic drug products pose a risk to patient safety. We investigated the threshold of biotherapeutic aggregates needed to induce immunogenic responses. METHODS AND RESULTS: Highly aggregated samples were tested in cell-based assays and induced cellular responses in a manner that depended on the number of particles. The threshold of immune activation varied by disease state (cancer, rheumatoid arthritis, allergy), concomitant therapies, and particle number. Compared to healthy donors, disease state patients showed an equal or lower response at the late phase (7 days), suggesting they may not have a higher risk of responding to aggregates. Xeno-het mice were used to assess the threshold of immune activation in vivo. Although highly aggregated samples (~ 1,600,000 particles/mL) induced a weak and transient immunogenic response in mice, a 100-fold dilution of this sample (~ 16,000 particles/mL) did not induce immunogenicity. To confirm this result, subvisible particles (up to ~ 18,000 particles/mL, containing aggregates and silicone oil droplets) produced under representative administration practices (created upon infusion of a drug product through an IV catheter) did not induce a response in cell-based assays or appear to increase the rate of adverse events or immunogenicity during phase 3 clinical trials. CONCLUSION: The ability of biotherapeutic aggregates to elicit an immune response in vitro, in vivo, and in the clinic depends on high numbers of particles. This suggests that there is a high threshold for aggregates to induce an immunogenic response which is well beyond that seen in standard biotherapeutic drug products.


Subject(s)
Antibody Formation , Humans , Mice , Animals , Pharmaceutical Preparations
2.
J Biotechnol ; 317: 5-15, 2020 Jun 20.
Article in English | MEDLINE | ID: mdl-32361021

ABSTRACT

Therapeutic monoclonal antibodies (mAbs), particularly of the IgG1 subclass, are capable of effector function activities that may be important for their mechanism of action. One such effector function activity is Antibody Dependent Cellular Phagocytosis (ADCP), which has been shown to be mediated primarily through the activating FcγR, FcγRIIa, on macrophages and neutrophils. The critical quality attributes that are the most impactful and predictive of ADCP activity, and therefore most suitable to monitor during IgG1 antibody manufacturing, are not well established. Primary cell assays for ADCP are often laborious and subject to donor to donor variability, making such assays less desirable for product characterization. By developing and employing an ADCP reporter gene assay, we have been able to determine with high sensitivity the glycan structures that can impact FcγRIIa mediated ADCP across multiple different IgG1 antibodies. Interestingly we observed that some IgG1 antibodies are very potent mediators of ADCP while others do not mediate ADCP even though they possess other effector function activities (ADCC and CDC). Additionally, we find that ADCP by different IgG1 antibodies has markedly different sensitivity to glycan species, with one antibody demonstrating a surprisingly strong influence of ß-galactosylation and high mannose levels.


Subject(s)
Antibodies, Monoclonal , Cytotoxicity, Immunologic/physiology , Phagocytosis/physiology , Polysaccharides/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , CHO Cells , Cricetinae , Cricetulus , Glycosylation , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Jurkat Cells , Receptors, IgG/chemistry , Receptors, IgG/metabolism
3.
Drug Metab Lett ; 10(2): 144-50, 2016.
Article in English | MEDLINE | ID: mdl-27165340

ABSTRACT

BACKGROUND: The study of novel sites of metabolism is important in understanding new mechanisms of biotransformation of a particular moiety by metabolic enzymes. This information is valuable in designing metabolically-stable compounds with drug-like properties. It may also provide insights into the existence of active and reactive metabolites. METHODS: We utilized small scale incubations to generate adequate amounts of the metabolite of interest. After purification, LC-MS/MS and Proton Nuclear Magnetic Resonance (1H-NMR) were utilized to unequivocally assign the novel site of glutathione conjugation on the purine ring system. RESULTS: A proposed novel site of glutathione conjugation was investigated on a diaminopurine-containing molecule. It was demonstrated that the formation of the glutathione conjugate at the C-6 position of the purine ring system was due to the bioactivation of the compound to a di-imine intermediate by CYP3A4, followed by the nucleophilic addition of glutathione. CONCLUSION: S-glutathionylation at C-6 position of a purine was proven unequivocally. This previously unreported mechanism constitutes a novel biotransformation for purines.


Subject(s)
Chromatography, Liquid/methods , Cytochrome P-450 CYP3A/metabolism , Glutathione/metabolism , Purines/metabolism , Animals , Dogs , Haplorhini , Humans , Magnetic Resonance Spectroscopy/methods , Mice , Rats , Tandem Mass Spectrometry
4.
Xenobiotica ; 43(5): 399-408, 2013 May.
Article in English | MEDLINE | ID: mdl-23046389

ABSTRACT

1. Aldehyde oxidase (AO) is a cytosolic enzyme that contributes to the Phase I metabolism of xenobiotics in human and preclinical species. 2. Current studies explored in vitro metabolism of zoniporide in various animal species and humans using S9 fractions. The animal species included commonly used pharmacology and toxicology models and domestic animals such as the cat, cow or bull, pig and horse. 3. In addition, gender and strain differences in some species were also explored. 4. All animals except the dog and cat converted zoniporide to 2-oxozoniporide (M1). 5. Michael-Menten kinetic studies were conducted in species that turned over zoniporide to M1. 6. Marked differences in KM, Vmax and Clint were observed in the oxidation of zoniporide. 7. Although the KM and Vmax of zoniporide oxidation in male and female human S9 was similar, some gender difference was observed in animals especially, in Vmax. 8. The domestic animals also showed marked species differences in the AO activity and affinity toward zoniporide.


Subject(s)
Aldehyde Oxidase/metabolism , Guanidines/metabolism , Pyrazoles/metabolism , Xenobiotics/metabolism , Animals , Cats , Dogs , Female , Guanidines/pharmacokinetics , Humans , Male , Pyrazoles/pharmacokinetics , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Species Specificity
5.
Drug Metab Dispos ; 40(8): 1575-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22587988

ABSTRACT

Current studies explored the effect of structural changes on the aldehyde oxidase (AO)-mediated metabolism of zoniporide (1). Zoniporide analogs with modifications of the acylguanidine moiety, the cyclopropyl group on the pyrazole ring, and the quinoline ring were studied for their AO-catalyzed metabolism using the human S9 fraction. Analysis of the half-lives suggested that subtle changes in the structure of 1 influenced its metabolism and that the guanidine and the quinoline moieties were prerequisites for AO-catalyzed oxidation to 2-oxozoniporide (M1). In contrast, replacement of the cyclopropyl group with other alkyl groups was tolerated. The effect of structural variation on AO properties was rationalized by docking 1 and its analogs into the human AO homology model. These studies indicated the importance of electrostatic, π-π stacking and hydrophobic interactions of the three motifs with residues in the active site. Differences in substrate properties were also rationalized by comparing their half-lives with cLogD, electrophilicity parameters [electrostatic potential (ESP) charges and energy of lowest unoccupied molecular orbitals (E(LUMO))], and the energies of formation of tetrahedral intermediates (J Med Chem 50:4642-4647, 2007). Whereas the success of energetics in predicting the AO substrate properties of analogs was 87%, the predictive ability of other descriptors was none (cLogD) to 60% (ESP charges and E(LUMO)). Overall, the structure-metabolism relationship could be rationalized using a combination of both the energy calculations and docking studies. This combination method can be incorporated into a strategy for mitigating AO liabilities observed in the lead candidate or studying structure-metabolism relationships of other AO substrates.


Subject(s)
Aldehyde Oxidase/metabolism , Guanidines/metabolism , Pyrazoles/metabolism , Biocatalysis , Chromatography, High Pressure Liquid , Half-Life , Kinetics , Models, Molecular , Oxidation-Reduction
6.
Exp Eye Res ; 93(3): 256-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21376717

ABSTRACT

Better control of intraocular pressure (IOP) is the most effective way to preserve visual field function in glaucomatous patients. While prostaglandin FP analogs are leading the therapeutic intervention for glaucoma, new target classes also are being identified with new lead compounds being developed for IOP reduction. One target class currently being investigated includes the prostaglandin EP receptor agonists. Recently PF-04217329 (Taprenepag isopropyl), a prodrug of CP-544326 (active acid metabolite), a potent and selective EP(2) receptor agonist, was successfully evaluated for its ocular hypotensive activity in a clinical study involving patients with primary open angle glaucoma. In the current manuscript, the preclinical attributes of CP-544326 and PF-0421329 have been described. CP-544326 was found to be a potent and selective EP(2) agonist (IC(50) = 10 nM; EC(50) = 2.8 nM) whose corneal permeability and ocular bioavailability were significantly increased when the compound was dosed as the isopropyl ester prodrug, PF-04217329. Topical ocular dosing of PF-04217329 was well tolerated in preclinical species and caused an elevation of cAMP in aqueous humor/iris-ciliary body indicative of in vivo EP(2) target receptor activation. Topical ocular dosing of PF-04217329 resulted in ocular exposure of CP-544326 at levels greater than the EC(50) for the EP(2) receptor. PF-04217329 when dosed once daily caused between 30 and 50% IOP reduction in single day studies in normotensive Dutch-belted rabbits, normotensive dogs, and laser-induced ocular hypertensive cynomolgus monkeys and 20-40% IOP reduction in multiple day studies compared to vehicle-dosed eyes. IOP reduction was sustained from 6 h through 24 h following a single topical dose. In conclusion, preclinical data generated thus far appear to support the clinical development of PF-04217329 as a novel compound for the treatment of glaucoma.


Subject(s)
Acetates/pharmacology , Antihypertensive Agents/pharmacology , Disease Models, Animal , Glaucoma, Open-Angle/drug therapy , Glaucoma/drug therapy , Intraocular Pressure/drug effects , Prodrugs/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Sulfonamides/pharmacology , Acetates/pharmacokinetics , Administration, Topical , Animals , Antihypertensive Agents/pharmacokinetics , Aqueous Humor/metabolism , Biological Availability , Calcium/metabolism , Ciliary Body/metabolism , Cornea/metabolism , Cyclic AMP/metabolism , Dogs , Drug Evaluation, Preclinical , Glaucoma/metabolism , Humans , Iris/metabolism , Macaca fascicularis , Male , Ophthalmic Solutions/pharmacokinetics , Ophthalmic Solutions/pharmacology , Prodrugs/pharmacokinetics , Rabbits , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Sulfonamides/pharmacokinetics , Tonometry, Ocular
7.
Drug Metab Dispos ; 39(3): 539-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21156812

ABSTRACT

A recent X-ray crystal structure of a rabbit cytochrome P450 2B4 (CYP2B4)-ticlopidine complex indicated that the compound could be modeled with either the thiophene or chlorophenyl group oriented toward the heme prosthetic group. Subsequent NMR relaxation and molecular docking studies suggested that orientation with the chlorophenyl ring closer to the heme was the preferred one. To evaluate the predictive value of these findings, the oxidation of ticlopidine by reconstituted CYP2B4 was studied and compared with CYP2B6, in which the thiophene portion of the molecule likely orients toward the heme. In vitro incubation of ticlopidine with both enzymes yielded the same set of metabolites: 7-hydroxyticlopidine (M1), 2-oxoticlopidine (M2), 5-(2-chlorobenzyl)thieno[3,2-c]pyridin-5-ium metabolite (M3), 5-(2-chlorobenzyl)thieno[3,2-c]pyridin-5-ium metabolite (M4), ticlopidine N-oxide (M5), and ticlopidine S-oxide dimer, a dimerization product of ticlopidine S-oxide (M6). The rates of metabolite formation deviated markedly from linearity with time, consistent with the known inactivation of CYP2B6 by ticlopidine. Fitting to a first-order equation yielded similar rate constants (k(obs)) for both enzymes. However, the amplitude (R(max)) of M1 and M6 formation was 4 to 5 times higher for CYP2B6 than CYP2B4, indicating a greater residence time of ticlopidine with its thiophene ring closer to heme in CYP2B6. In contrast, CYP2B4 formed M4 and M5 in more abundance than CYP2B6, indicating an alternate orientation. Overall, the results suggest that the preferential orientation of ticlopidine in the active site of CYP2B4 predicted by X-ray crystallography and NMR studies is unproductive and that ticlopidine likely reorients within CYP2B4 to a more productive mode.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 Enzyme System/metabolism , Platelet Aggregation Inhibitors/metabolism , Ticlopidine/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/isolation & purification , Biocatalysis , Bupropion/metabolism , Chromatography, High Pressure Liquid , Coumarins/metabolism , Cytochrome P-450 CYP2B6 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/isolation & purification , Cytochrome P450 Family 2 , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Models, Molecular , Molecular Structure , Oxidation-Reduction , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Rabbits , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Ticlopidine/analogs & derivatives , Ticlopidine/chemistry , Ticlopidine/pharmacology
8.
J Pharmacol Exp Ther ; 331(2): 627-35, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19690190

ABSTRACT

Developing a population-based pharmacokinetic-pharmacodynamic (PKPD) model is a challenge in ophthalmology due to the difficulty of obtaining adequate pharmacokinetic (PK) samples from ocular tissues to inform the pharmacodynamic (PD) model. Using limited PK data, we developed a preclinical population-based PD model suitable for capturing the time course of dog intraocular pressure (IOP) that exhibited time-dependent sensitization after topical administration of PF-04475270 [5-{3-[(2S)-2-{(3R)-3-hydroxy-4-[3-(trifluoromethyl)phenyl]butyl}-5-oxopyrrolidin-1-yl]propyl}thiophene-2-carboxylate]. A physiologically relevant PK model was chosen to simultaneously capture the concentration profiles of CP-734432, a potent EP4 agonist and the active metabolite of PF-04475270, sampled from three ocular tissues of the anterior chamber: cornea, aqueous humor, and iris-ciliary body. Two population-based PD models were developed to characterize the IOP lowering profiles: model I, a standard indirect-response model (IRM); and model II, an extension of a standard IRM that empirically incorporated a response-driven positive feedback loop to account for the observed PD sensitization. The PK model reasonably described the PK profiles in all three ocular tissues. As for the PD, model I failed to capture the overall trend in the population IOP data, and model II more adequately characterized the overall data set. This integrated PKPD model may have general utility when PD sensitization is observed and is not a result of time-dependent PK. In addition, the model is applicable in the ophthalmology drug development setting in which PK information is limited but a population-based PD model could reasonably be established.


Subject(s)
Intraocular Pressure/drug effects , Ocular Hypertension/drug therapy , Pyrrolidinones/pharmacology , Receptors, Prostaglandin E/agonists , Thiophenes/pharmacology , Algorithms , Animals , Aqueous Humor/metabolism , Ciliary Body/metabolism , Dogs , Female , Glaucoma/metabolism , Humans , Male , Models, Statistical , Ocular Hypertension/physiopathology , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Pyrrolidinones/therapeutic use , Receptors, Prostaglandin E, EP4 Subtype , Thiophenes/therapeutic use
9.
J Ocul Pharmacol Ther ; 25(3): 215-22, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19456256

ABSTRACT

PURPOSE: Steroids are used in a diverse range of conditions in clinical ophthalmology and one of the most significant complications is corticosteroid-induced glaucoma, which is characterized by an increase in intraocular pressure (IOP). 11beta-Hydroxysteroid dehydrogenase-1 (11beta-HSD1) is known to catalyze the interconversion of hormonally inactive cortisone to hormonally active cortisol and is widely expressed in the eye, particularly ciliary epithelium. Carbenoxolone (CBX), an 11beta-HSD1 inhibitor, has been shown to reduce IOP in healthy volunteers and patients with ocular hypertension (OHT). The purpose of this study was to: (1) develop an in vivo model for the assessment of cortisone to cortisol conversion in the eye, that is, 11beta-HSD1 activity and (2) assess the pharmacokinetic/pharmacodynamic relationship following topical treatment with 11beta-HSD1 inhibitors using an in vivo rabbit model. METHODS: Potent and selective 11beta-HSD1 inhibitors were topically administered to the rabbit eye and exogenous cortisone to endogenous cortisol conversion in the eye was assessed in rabbits. Tissues were then evaluated for cortisone, cortisol, and 11beta-HSD1 inhibitor levels by LC/MS/MS. Concomitantly cortisol activity in ocular tissue samples was determined using a secondary mechanistic pLuc-GRE assay. RESULTS: Topical treatment with potent and selective 11beta-HSD1 inhibitors resulted in complete inhibition in the conversion of cortisone to cortisol in the rabbit eye as well as decreased pLuc-GRE luciferase activity. The reduction of cortisone conversion was time- and dose-dependent as well as dependent on dosing volume (suggestive of increased spillover and washout with greater dosing volume). CONCLUSIONS: In conclusion, topical delivery of 11beta-HSD1 inhibitors can reduce or inhibit the conversion of cortisone to cortisol in the eye, indicating that the rabbit eye possesses an active enzyme for glucocorticoid synthesis. Dosing concentration and volume play an important role in the pharmacokinetic and pharmacodynamic effects of topically delivering an 11beta-HSD1 inhibitor. The rabbit model is useful for mechanistically assessing the conversion of cortisone to cortisol in the eye.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Eye/drug effects , Intraocular Pressure/drug effects , Administration, Topical , Animals , Aqueous Humor/chemistry , Chromatography, Liquid , Cortisone/metabolism , Dose-Response Relationship, Drug , Eye/metabolism , Glucocorticoids/metabolism , Hydrocortisone/metabolism , Rabbits , Tandem Mass Spectrometry
10.
Exp Eye Res ; 89(5): 608-17, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19445930

ABSTRACT

Prostaglandins are widely used to lower intraocular pressure (IOP) as part of the treatment regimen for glaucoma. While FP and EP2 agonists are known to lower IOP, we investigated the ocular hypotensive activity and ocular drug distribution of PF-04475270, a novel EP4 agonist following topical administration in normotensive Beagle dogs. PF-04475270 is a prodrug of CP-734432, which stimulated cAMP formation in HEK293 cells expressing EP4 receptor and beta-lactamase activity in human EP4 expressing CHO cells transfected with a cAMP response element (CRE) with an EC(50) of 1 nM. Prodrug conversion and transcorneal permeability were assessed in rabbit corneal homogenates and a human corneal epithelial cell (cHCE) model. The compound underwent rapid hydrolysis to CP-734432 in corneal homogenates, and exhibited good permeability in the cHCE model. The descending order of ocular exposure to CP-734432 after topical dosing of PF-04475270 in dogs was as follows: cornea > aqueous humor >or= iris/ciliary body. When administered q.d., PF-04475270 lowered IOP effectively in the dog IOP model both after single and multiple days of dosing. A maximum decrease in IOP with PF-04475270 was between 30 and 45% at 24h post-dose relative to that observed with vehicle. In conclusion, PF-04475270 is a novel ocular hypotensive compound which is bioavailable following topical dosing, effectively lowering IOP in dogs. EP4 agonists could be considered as potential targets for lowering IOP for the treatment of glaucoma and ocular hypertension.


Subject(s)
Eye/drug effects , Intraocular Pressure/drug effects , Prodrugs/pharmacokinetics , Pyrrolidinones/pharmacokinetics , Receptors, Prostaglandin E/agonists , Thiophenes/pharmacokinetics , Administration, Topical , Animals , Biological Availability , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Dogs , Dose-Response Relationship, Drug , Eye/metabolism , Eye/pathology , Humans , Hydrolysis , Hyperemia/chemically induced , Models, Animal , Ophthalmic Solutions , Permeability , Prodrugs/administration & dosage , Prodrugs/toxicity , Pyrrolidinones/administration & dosage , Pyrrolidinones/toxicity , Rabbits , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP4 Subtype , Thiophenes/administration & dosage , Thiophenes/toxicity , Transfection
11.
Drug Metab Dispos ; 37(5): 992-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19220984

ABSTRACT

The recently introduced Clonetics human corneal epithelium (cHCE) cell line is considered a promising in vitro permeability model, replacing excised animal cornea to predict corneal permeability of topically administered compounds. The purpose of this study was to further characterize cHCE as a corneal permeability model from both drug metabolism and transport aspects. First, good correlation was found in the permeability values (P(app)) obtained from cHCE and rabbit corneas for various ophthalmic drugs and permeability markers. Second, a previously established real-time quantitative polymerase chain reaction method was used to profile mRNA expression of drug-metabolizing enzymes (major cytochromes P450 and UDP glucuronosyltransferase 1A1) and transporters in cHCE in comparison with human cornea. Findings indicated that 1) the mRNA expression of most metabolizing enzymes tested was lower in cHCE than in excised human cornea, 2) the mRNA expression of efflux transporters [multidrug resistant-associated protein (MRP) 1, MRP2, MRP3, and breast cancer resistance protein], peptide transporters (PEPT1 and PEPT2), and organic cation transporters (OCTN1, OCTN2, OCT1, and OCT3) could be detected in cHCE as in human cornea. However, multidrug resistance (MDR) 1 and organic anion transporting polypeptide 2B1 was not detected in cHCE; 3) cHCE was demonstrated to possess both esterase and ketone reductase activities known to be present in human cornea; and 4) transport studies using probe substrates suggested that both active efflux and uptake transport may be limited in cHCE. As the first detailed report to delineate drug metabolism and transport characteristics of cHCE, this work shed light on the usefulness and potential limitations of cHCE in predicting the corneal permeability of ophthalmic drugs, including ester prodrugs, and transporter substrates.


Subject(s)
Cornea/cytology , Cornea/metabolism , Epithelial Cells/metabolism , Alcohol Oxidoreductases/metabolism , Animals , Cell Line , Chromatography, High Pressure Liquid , Cornea/enzymology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Digoxin/metabolism , Epithelial Cells/enzymology , Esterases/metabolism , Humans , Indinavir/metabolism , Mass Spectrometry , Permeability , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rabbits , Reverse Transcriptase Polymerase Chain Reaction
12.
Drug Metab Dispos ; 36(7): 1300-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18411399

ABSTRACT

Studies were designed to quantitatively assess the mRNA expression of 1) 10 cytochrome P450 (P450) enzymes in human cornea, iris-ciliary body (ICB), and retina/choroid relative to their levels in the liver, and of 2) 21 drug transporters in these tissues relative to their levels in human small intestine, liver, or kidney. Potential species differences in mRNA expression of PEPT1, PEPT2, and MDR1 were also assessed in these ocular tissues from rabbit, dog, monkey, and human. P450 expression was either absent or marginal in human cornea, ICB, and retina/choroid, suggesting a limited role for P450-mediated metabolism in ocular drug disposition. In contrast, among 21 key drug efflux and uptake transporters, many exhibited relative expression levels in ocular tissues comparable with those observed in small intestine, liver, or kidney. This robust ocular transporter presence strongly suggests a significant role that transporters may play in ocular barrier function and ocular pharmacokinetics. The highly expressed efflux transporter MRP1 and uptake transporters PEPT2, OCT1, OCTN1, and OCTN2 may be particularly important in absorption, distribution, and clearance of their drug substrates in the eye. Evidence of cross-species ocular transporter expression differences noted in these studies supports the conclusion that transporter expression variability, along with anatomic and physiological differences, should be taken into consideration to better understand animal ocular pharmacokinetic and pharmacodynamic data and the scalability to human for ocular drugs.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Eye/metabolism , Pharmaceutical Preparations/metabolism , RNA, Messenger/genetics , Animals , Base Sequence , DNA Primers , Dogs , Female , Haplorhini , Humans , Male , Pharmacokinetics , Polymerase Chain Reaction , Rabbits
13.
Chem Res Toxicol ; 21(12): 2260-71, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19548350

ABSTRACT

Raloxifene (Evista) is a second generation selective estrogen receptor modulator used in the treatment of osteoporosis and for chemoprevention of breast cancer. It is bioactivated to reactive intermediates, which covalently bind to proteins and form GSH conjugates upon incubation with NADPH and GSH-supplemented human and rat liver microsomes. Despite these in vitro findings, no major raloxifene-related toxic events have been reported upon its oral administration to humans. This disconnect between safety of raloxifene and its in vitro bioactivation is attributed to its presystemic metabolism via glucuronidation. Current studies investigated the effect of hepatic and intestinal glucuronidation in modulating hepatic availability of raloxifene and its subsequent bioactivation, in vitro. The study design involved preincubation of raloxifene with intestinal microsomes followed by a sequential incubation with liver microsomes. The degree of bioactivation of raloxifene was assessed from the percentage of GSH conjugate formed in liver microsomal incubations or the amount of covalent binding of raloxifene-related material to liver microsomal proteins. The results indicated that human intestinal glucuronidation limited the hepatic exposure of raloxifene that underwent bioactivation in the liver. Similar experiments with rat microsomal preparations showed very little effect of intestinal glucuronidation. This effect of intestinal glucuronidation and the observed species difference were explained by comparing the efficiency (Cl(int)) of glucuronidation and oxidation in the two species. These findings suggested that even though the rate of bioactivation in the two species was similar, the Cl(int) of glucuronidation was 7.5-fold higher in the human intestine as compared to rats. These results support the hypothesis that intestinal glucuronidation modulates the amount of raloxifene undergoing bioactivation by liver and corroborate the importance of assessing other competitive metabolic pathways and species differences in metabolism prior to extrapolation of bioactivation results from rats to humans.


Subject(s)
Bone Density Conservation Agents/pharmacokinetics , Glucuronides/metabolism , Intestinal Mucosa/metabolism , Liver/metabolism , Microsomes, Liver/metabolism , Raloxifene Hydrochloride/pharmacokinetics , Animals , Biotransformation , Bone Density Conservation Agents/metabolism , Humans , Intestines/drug effects , Liver/drug effects , Microsomes, Liver/drug effects , Raloxifene Hydrochloride/metabolism , Rats , Species Specificity
14.
Curr Drug Metab ; 8(2): 91-107, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17305490

ABSTRACT

Ophthalmic drugs are delivered to ocular tissues predominantly via relatively simple formulations, such as topically dosed water-soluble drug solutions and water-insoluble drug suspensions in ointments. An ideal topical drug delivery system should possess certain desirable properties, such as good corneal and conjunctival penetration, prolonged precorneal residence time, easy instillation, non-irritative and comfortable to minimize lachrymation and reflex blinking, and appropriate rheological properties. In general, ocular efficacy is closely related to ocular drug bioavailability, which may be enhanced by increasing corneal drug penetration and prolonging precorneal drug residence time. To improve ocular bioavailability of topically dosed ophthalmic drugs, a variety of ocular drug delivery systems, such as hydrogels, microparticles, nanoparticles, microemulsions, liposomes and collagen shields, have been designed and investigated. These newer systems may, to some extent, control drug release and maintain therapeutic levels in ocular tissues over a prolonged period of time. This review focuses on the in vitro, ex vivo and in vivo studies of ophthalmic drugs formulated in nanoparticles published over the past two decades. The progress and development issues relating to ocular disposition, pharmacokinetics, efficacy and safety of the nanoparticle-formulated ophthalmic drugs are specifically addressed. Information and discussions summarized in this review are helpful for pharmaceutical scientists to develop better ophthalmic therapeutics.


Subject(s)
Drug Delivery Systems , Eye/metabolism , Nanoparticles , Animals , Humans , Nanoparticles/adverse effects , Nanoparticles/therapeutic use , Ophthalmic Solutions/adverse effects , Ophthalmic Solutions/pharmacokinetics , Ophthalmic Solutions/therapeutic use , Polymers/adverse effects , Polymers/pharmacokinetics , Polymers/therapeutic use , Treatment Outcome
15.
Eur J Pharm Sci ; 26(2): 151-61, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16085400

ABSTRACT

Enterohepatic recirculation (EHR) occurs via biliary excretion and intestinal reabsorption of a drug. Drug recycling through EHR can lead to a change in pharmacokinetic (PK) properties, such as reduced clearance (CL), extended half-life (T(1/2)) and increased plasma exposure (AUC). As a result, EHR may prolong the pharmacological effect of drugs. In the present study, the compound (Cpd A) was found to exhibit EHR in Rhesus monkeys associated with a reduction in CL (from 3.8 to 0.33 Lh(-1), IV; from 2.3 to 0.4 Lh(-1), PO), and an increase in T(1/2) (from 0.9 to 18 h, IV) and in AUC (from 1.5 to 17.4 microg h/mL, IV; from 2.8 to 16.3 microg h/mL, PO), by comparing the PK in the monkeys via the interruption of EHR (bile-duct cannulation) with that in the intact monkeys. A population four-compartment model was constructed based on recirculation loops incorporating all possible inputs (bile secretion, a lag-time model for gall bladder emptying, routes and amounts of a single dose administration) to fully evaluate the EHR of Cpd A. The plasma concentrations versus time profiles predicted from the model had a good fit to the values observed in the subjects and were further simulated with 90% confidence interval to demonstrate its utility. Thus, the model could be applied as a useful tool to evaluate the drugs or compounds that undergo EHR in different species.


Subject(s)
Bile/metabolism , Enterohepatic Circulation , Models, Biological , Pharmacokinetics , Animals , Area Under Curve , Bile/chemistry , Biotransformation , Common Bile Duct/surgery , Half-Life , Intestinal Absorption , Liver/metabolism , Macaca mulatta , Male , Metabolic Clearance Rate , Xenobiotics/pharmacokinetics , Xenobiotics/pharmacology
16.
J Pharmacol Exp Ther ; 313(2): 518-28, 2005 May.
Article in English | MEDLINE | ID: mdl-15677349

ABSTRACT

In the present study, N-(alpha-methylbenzyl-)-1-aminobenzotriazole (MBA) and ketoconazole (KET) were identified as the inhibitors with selectivity toward dog CYP2B11 and CYP3A12, respectively. Their selectivity was evaluated using phenacetin O-deethylation (CYP1A), diazepam (DZ) N1-demethylation (CYP2B11), diclofenac 4'-hydrxylation (CYP2C21), bufuralol 1'-hydroxylation (CYP2D11), and DZ C3-hydroxylation (CYP3A12) activities in dog liver microsomes (DLM). MBA exhibited potent mechanism-based inhibition of DZ N1-demethylase activity catalyzed by both baculovirus-expressed CYP2B11 and DLM. In both cases, inhibition was characterized by a low K(I) (0.35 and 0.46 microM, respectively) and high k(inact) (1.5 and 0.56 min(-1), respectively). Despite complete loss of DZ N1-demethylase activity in the presence of MBA, there was no significant loss of cytochrome P450 (P450) CO-binding spectrum. These data suggest that the inactivation involved covalent modification of P450 apoprotein, instead of the prosthetic heme moiety. A homology model of CYP2B11 was constructed, based on the crystal structure of rabbit CYP2C5, for docking the substrate (DZ) and the inhibitor (MBA), respectively. The model, within the limits of our approximations, helped explain the substrate specificity and inhibitor selectivity of CYP2B11. In contrast to MBA, KET was identified as a potent and selective reversible (competitive) inhibitor of CYP3A12 (K(I) = 0.13-0.33 microM). In fact, complete inhibition of CYP3A12-dependent DZ C3-hydroxylation was possible at a low KET concentration (1 microM). Therefore, it is concluded that one can attempt to conduct P450 reaction phenotype studies with DLM using MBA and KET as selective inhibitors of CYP2B11 and CYP3A12, respectively.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Cytochrome P-450 Enzyme Inhibitors , Liver/enzymology , Steroid Hydroxylases/antagonists & inhibitors , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Binding Sites/drug effects , Binding Sites/physiology , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 2 , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ketoconazole/pharmacology , Liver/drug effects , Steroid Hydroxylases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...