Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
FASEB J ; 31(1): 368-375, 2017 01.
Article in English | MEDLINE | ID: mdl-27770021

ABSTRACT

The purpose of these studies was to test the hypothesis that a selected polypharmacological approach for treating the prostanoid-mediated component of inflammatory diseases would produce a therapeutic effect superior to global inhibition of prostaglandin (PG) biosynthesis by aspirin-like drugs. The compound studied was AGN 211377, which had been previously shown to produce a superior effect on cytokine release from human macrophages compared with cyclooxygenase (COX) inhibitors. AGN 211377 antagonizes prostanoid prostaglandin D2 (DP)1, DP2, prostaglandin E2 (EP)1, EP4, prostaglandin F2α, and thromboxane A2 receptors but not anti-inflammatory EP2, prostaglandin I2, or EP3 receptors. Established rodent models of ocular inflammatory diseases were used to determine therapeutic effects in living animals. The drugs were administered systemically after predetermination of their blood levels to ensure bioavailability at an appropriate dose level. Whereas compounds selective for a single prostanoid receptor typically exhibited modest but statistically significant inhibition, AGN 211377 profoundly inhibited S-antigen-induced uveitis and laser-induced retinal neovascularization. Consistent with previous polypharmacological studies on chemokine/cytokine release from human macrophages, the prostanoid EP1 receptor played a permissive role in suppressing neovascularization and inflammation in vivo Comparing AGN 211377 with a close structural congener lacking EP1 antagonism (AGN 197727), AGN 197727 was much less active than AGN 211377, but pronounced anti-inflammatory and angiostatic effects were achieved by adding the EP1 antagonist compound (SC-51322) to AGN 197727 in the systemic dosing regimen. Further, AGN 211377 produced superior anti-inflammatory activity compared with the nonsteroidal anti-inflammatory agent ketorolac. These results indicate the value of using a polypharmacological approach in the design of novel therapeutic agents in preference to compounds targeting a single receptor or enzyme. A compound such as AGN 211377 may represent more effective therapy than COX inhibitors in treating uveitis and ocular diseases where neovascularization is a significant part of the pathology.-Woodward, D. F., Wang, J. W., Ni, M., Bauer, A., Martos, J. L., Carling, R. W., Poloso, N. J. In vivo studies validating multitargeting of prostanoid receptors for achieving superior anti-inflammatory effects.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cinnamates/pharmacology , Neovascularization, Pathologic/prevention & control , Receptors, Prostaglandin/antagonists & inhibitors , Animals , Autoimmune Diseases , Calcium Signaling , Lasers/adverse effects , Rats , Rats, Inbred Lew , Retina/pathology , Retina/radiation effects , Retinal Vessels/pathology , Retinal Vessels/radiation effects , Uveitis/drug therapy , Uveitis/etiology
2.
FASEB J ; 30(1): 394-404, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26420849

ABSTRACT

A polypharmacologic approach to prostanoid based anti-inflammatory therapeutics was undertaken in order to exploit both the anti- and proinflammatory properties attributed to the various prostanoid receptors. Multitargeting of selected prostanoid receptors yielded a prototype compound, compound 1 (AGN 211377), that antagonizes prostaglandin D2 receptors (DPs) DP1 (49) and DP2 (558), prostaglandin E2 receptors (EPs) EP1 (266) and EP4 (117), prostaglandin F2α receptor (FP) (61), and thromboxane A2 receptor (TP) (11) while sparing EP2, EP3, and prostaglandin I2 receptors (IPs); Kb values (in nanomoles) are given in parentheses. Compound 1 evoked a pronounced inhibition of cytokine/chemokine secretion from lipopolysaccharide or TNF-α stimulated primary human macrophages. These cytokine/chemokines included cluster of designation 40 receptor (CD40), epithelial-derived neutrophil-activating protein 78 (ENA-78), granulocyte colony stimulating factor (G-CSF), granulocyte macrophage colony stimulating factor (GM-CSF), IL-8, IL-18, monocyte chemotactic protein-1 (CCL2) (MCP-1), tissue plasminogen activator inhibitor (PAI-1), and regulated on activation, normal T cell expressed and secreted (RANTES). In contrast, the inhibitory effects of most antagonists selective for a single receptor were modest or absent, and selective EP2 receptor blockade increased cytokine release in some instances. Compound 1 also showed clear superiority to the cyclooxygenase inhibitors diclofenac and rofecoxib. These findings reveal that blockade of multiple prostanoid receptors, with absent antagonism of EP2 and IP, may provide more effective anti-inflammatory activity than global suppression of prostanoid synthesis or highly selective prostanoid receptor blockade. These investigations demonstrate the first working example of prostanoid receptor polypharmacology for potentially safer and more effective anti-inflammatory therapeutics by blocking multiple proinflammatory receptors while sparing those with anti-inflammatory activity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cinnamates/pharmacology , Macrophages/drug effects , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Thromboxane/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Cells, Cultured , Cinnamates/chemical synthesis , Humans , Macrophages/metabolism , Substrate Specificity
3.
J Pharmacol Exp Ther ; 358(2): 173-80, 2016 08.
Article in English | MEDLINE | ID: mdl-27217589

ABSTRACT

Prostaglandin E2 (PGE2)-2-glyceryl ester is a cyclo-oxygenase 2 product of the endocannabinoid 2-arachidonyl glycerol. It is claimed as pharmacologically novel, but this is complicated by rapid and irreversible isomerization to the 1(3) ester. For ocular studies, enzymatic hydrolysis of the ester moiety creates an additional complication. PG-glyceryl esters were stabilized to isomerization and hydrolysis by replacing the noncarbonyl O with NH, to form the serinolamide and propanediolamide as stable analogs of PG-2-glyceryl and PG-2-1(3) glyceryl esters, respectively. Intraocular pressure was measured in conscious dogs and conscious laser-induced ocular hypertensive monkeys. Pharmacological studies involved stable transfectants for each of the human recombinant prostanoid receptors and the isolated feline iris for prostamide activity. PGE2-serinolamide and PGE2- propanediolamide were essentially inactive at all receptors except the EP3 receptor (EC50, ∼500 nM). This obliged elucidation of EP3 receptor involvement in the intraocular pressure response to these PGE2-glycyerl ester analogs. Since the EP3 receptor agonists sulprostone and GR 63799 did not lower monkey intraocular pressure, a role for EP3 receptors in mediating the effects of PGE2-serinolamide and PGE2-propanediolamide is not indicated. PGE2-glyceryl ester (0.01% and 0.1%) substantially lowered intraocular pressure in monkeys. PGE2-propanediolamide was more efficacious than PGE2-serinolamide in lowering intraocular pressure in monkey eyes, but both appeared equieffective in dog eyes. PGE2-serinolamide dose-dependently (0.01- 0.1%) lowered intraocular pressure in both species, but PGF2 α-serinolamide was inactive. In conclusion, stable PGE2-glyceryl ester analogs lowered intraocular pressure. These findings are consistent with the presence of a PGE2-glyceryl ester-specific recognition site in the eye.


Subject(s)
Dinoprostone/analogs & derivatives , Intraocular Pressure/drug effects , Animals , Cats , Dinoprostone/pharmacology , Dogs , Female , Iris/drug effects , Iris/physiology , Macaca fascicularis , Male , Muscle Contraction/drug effects
4.
FASEB J ; 27(2): 557-67, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23104985

ABSTRACT

Balding causes widespread psychological distress but is poorly controlled. The commonest treatment, minoxidil, was originally an antihypertensive drug that promoted unwanted hair. We hypothesized that another serendipitous discovery, increased eyelash growth side-effects of prostamide F(2α)-related eyedrops for glaucoma, may be relevant for scalp alopecias. Eyelash hairs and follicles are highly specialized and remain unaffected by androgens that inhibit scalp follicles and stimulate many others. Therefore, we investigated whether non-eyelash follicles could respond to bimatoprost, a prostamide F(2α) analog recently licensed for eyelash hypotrichosis. Bimatoprost, at pharmacologically selective concentrations, increased hair synthesis in scalp follicle organ culture and advanced mouse pelage hair regrowth in vivo compared to vehicle alone. A prostamide receptor antagonist blocked isolated follicle growth, confirming a direct, receptor-mediated mechanism within follicles; RT-PCR analysis identified 3 relevant receptor genes in scalp follicles in vivo. Receptors were located in the key follicle regulator, the dermal papilla, by analyzing individual follicular structures and immunohistochemistry. Thus, bimatoprost stimulates human scalp follicles in culture and rodent pelage follicles in vivo, mirroring eyelash behavior, and scalp follicles contain bimatoprost-sensitive prostamide receptors in vivo. This highlights a new follicular signaling system and confirms that bimatoprost offers a novel, low-risk therapeutic approach for scalp alopecias.


Subject(s)
Alopecia/drug therapy , Amides/therapeutic use , Cloprostenol/analogs & derivatives , Administration, Topical , Adult , Alopecia/genetics , Alopecia/metabolism , Amides/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/therapeutic use , Base Sequence , Bimatoprost , Cloprostenol/administration & dosage , Cloprostenol/therapeutic use , Female , Glaucoma/drug therapy , Hair Follicle/drug effects , Hair Follicle/growth & development , Hair Follicle/metabolism , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Organ Culture Techniques , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/drug effects , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Scalp/drug effects , Scalp/metabolism , Young Adult
5.
Exp Dermatol ; 22(2): 145-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23278986

ABSTRACT

Studies on bimatoprost were performed with two objectives: (i) to determine whether bimatoprost possesses hair growth-stimulating properties beyond eyelash hypertrichosis and (ii) to investigate the biodisposition of bimatoprost in skin for the first time. Bimatoprost, at the dose used clinically for eyelash growth (0.03%) and given once daily for 14 days, increased pelage hair growth in C57/black 6 mice. This occurred as a much earlier onset of new hair growth in shaved mice and the time taken to achieve complete hair regrowth, according to photographic documentation and visual assessment. Bimatoprost biodisposition in the skin was determined at three concentrations: 0.01%, 0.03% and 0.06%. Dose-dependent C(max) values were obtained (3.41, 6.74, 12.3 µg/g tissue), and cutaneous bimatoprost was well maintained for 24 h following a single dose. Bimatoprost was recovered from the skin only as the intact molecule, with no detectable levels of metabolites. Thus, bimatoprost produces hypertrichosis as the intact molecule.


Subject(s)
Amides/pharmacology , Amides/pharmacokinetics , Cloprostenol/analogs & derivatives , Hypertrichosis/chemically induced , Skin/drug effects , Animals , Antihypertensive Agents/pharmacology , Bimatoprost , Cloprostenol/pharmacokinetics , Cloprostenol/pharmacology , Dose-Response Relationship, Drug , Eyelashes , Hair/physiology , Hypertrichosis/pathology , Mice , Mice, Inbred C57BL , Prostaglandins/metabolism , Skin/pathology , Time Factors
6.
Br J Ophthalmol ; 105(11): 1610-1616, 2021 11.
Article in English | MEDLINE | ID: mdl-33239414

ABSTRACT

BACKGROUND: A single application of JV-GL1 substantially lowers non-human primate intraocular pressure (IOP) for about a week, independent of dose. This highly protracted effect does not correlate with its ocular biodisposition or correlate with the once-daily dosing regimen for other prostanoid EP2 receptor agonists such as trapenepag or omidenepag. The underlying pharmacological mechanism for the multiday extended activity of JV-GL1 is highly intriguing. The present studies were intended to determine EP2 receptor involvement in mediating the long-term ocular hypotensive activity of JV-GL1 by using mice genetically deficient in EP2 receptors. METHODS: The protracted IOP reduction produced by JV-GL1 was investigated in C57BL/6J and EP2 receptor knock-out mice (B6.129-Ptger2tm1Brey /J; EP2KO). Both ocular normotensive and steroid-induced ocular hypertensive (SI-OHT) mice were studied. IOP was measured tonometrically under general anaesthesia. Aqueous humour outflow facility was measured ex vivo using iPerfusion in normotensive C57BL/6J mouse eyes perfused with 100 nM de-esterified JV-GL1 and in SI-OHT C57BL/6J mouse eyes that had received topical JV-GL1 (0.01%) 3 days prior. RESULTS: Both the initial 1-day and the protracted multiday effects of JV-GL1 in the SI-OHT model for glaucoma were abolished by deletion of the gene encoding the EP2 receptor. Thus, JV-GL1 did not lower IOP in SI-OHT EP2KO mice, but in littermate SI-OHT EP2WT control mice, JV-GL1 statistically significantly lowered IOP for 4-6 days. CONCLUSIONS: Both the 1-day and the long-term effects of JV-GL1 on IOP are entirely EP2 receptor dependent.


Subject(s)
Intraocular Pressure , Ocular Hypertension , Ocular Hypotension , Animals , Antihypertensive Agents/therapeutic use , Intraocular Pressure/drug effects , Mice , Mice, Inbred C57BL , Ocular Hypertension/drug therapy , Ocular Hypotension/drug therapy , Ophthalmic Solutions/administration & dosage , Tonometry, Ocular
7.
Prostaglandins Other Lipid Mediat ; 92(1-4): 19-24, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20152925

ABSTRACT

2-Arachidonoylglycerol is oxygenated by cyclooxygenase-2 to form prostaglandin glyceryl esters. Previous work in this laboratory has suggested that PGE(2)-G activates a novel G protein-coupled receptor in a murine macrophage-like cell line, RAW 264.7. To probe the structural determinants for the putative receptor in RAW 264.7 cells, a panel of 10 analogs was tested for their ability to increase intracellular calcium. These analogs included PGE(2)- and PGF(2alpha)-ethanolamide, 4 PGE(2) glyceryl ester analogs, and 4 PGF(2alpha) glyceryl ester analogs. The glyceryl ester analogs differed in the positioning of the hydroxyl groups in the glycerol moiety and the type of linker (ester, amide, or thioester) of the prostaglandin to the glycerol moiety. Compounds were also evaluated in a human non-small cell lung cancer cell line (H1819). The glycerol moiety was required for the calcium response. All glyceryl ester analogs but not ethanolamides caused a concentration-dependent increase in calcium levels in both RAW 264.7 and H1819 cells. An amide or ester linkage was preferable to a thioester linkage. The EC(50) values did not significantly change when the positioning of the hydroxyls was varied. This calcium response induced by the glyceryl ester analogs appears to be independent of the putative hydrolysis products, PGE(2) and PGF(2alpha), and appears to represent a novel signaling pathway.


Subject(s)
Calcium Signaling/drug effects , Dinoprost/chemistry , Dinoprost/pharmacology , Dinoprostone/chemistry , Dinoprostone/pharmacology , Esters/chemistry , Animals , Cell Line, Tumor , Ethanolamine/chemistry , Humans , Mice , Structure-Activity Relationship
8.
J Ocul Pharmacol Ther ; 36(8): 636-648, 2020 10.
Article in English | MEDLINE | ID: mdl-32640926

ABSTRACT

Purpose: JV-GL1 is an efficacious, potent, and long-acting antiglaucoma agent, according to studies in ocular normotensive and hypertensive monkeys. As an obligatory step in the drug development process, studies with exaggerated doses and an accelerated dosing schedule for JV-GL1 were performed in a second species (dog). Methods: Intraocular pressure (IOP) was measured by pneumatonometry in conscious Beagle dogs, which remained conscious throughout the study and gently restrained by hand. Pupil diameter was measured with an Optistick. Ocular surface hyperemia was visually assessed and scored according to a 1-3 assessment scale. Results: JV-GL1, as a 0.01% eye drop, produced significantly greater reductions in IOP than the original clinical dose of bimatoprost (0.03%). JV-GL1 and its free acid enzymatic hydrolysis product PGN 9856, over a 0.01%-0.1% dose range, reduced IOP to ≤10 mm Hg. JV-GL1 and PGN 9856 produced no miosis but a similar degree of ocular surface hyperemia to bimatoprost. Although PGN 9862, a close congener of PGN 9856, was very active as the free acid, esterification essentially abolished its ocular hypotensive activity and ocular surface redness. Conclusion: JV-GL1 was confirmed as a highly effective and potent ocular hypotensive, exceeding the activity of bimatoprost. A similar degree of ocular surface redness was apparent for both compounds, given as eye drops, but no other effects occurred. Results with PGN 9862 and its isopropyl ester confirmed that PGN 9862-isopropyl ester is not bioavailable in the eye and not susceptible to enzymatic hydrolysis in ocular tissues, a first for C1 ester prodrugs in the eye.


Subject(s)
Acetates/pharmacology , Antihypertensive Agents/pharmacology , Biphenyl Compounds/pharmacology , Eye/drug effects , Glaucoma/drug therapy , Acetates/administration & dosage , Acetates/adverse effects , Acetates/chemistry , Administration, Topical , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/adverse effects , Antihypertensive Agents/chemistry , Bimatoprost/pharmacology , Biological Availability , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/adverse effects , Biphenyl Compounds/chemistry , Dogs , Drug Development/methods , Eye/metabolism , Eye/physiopathology , Female , Hydrolysis , Hyperemia/chemically induced , Intraocular Pressure/drug effects , Male , Models, Animal , Ophthalmic Solutions/administration & dosage , Safety
9.
ACS Pharmacol Transl Sci ; 3(6): 1199-1210, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33344897

ABSTRACT

It has previously been reported that a prototypical compound (AGN 211377), which blocks pro-inflammatory prostanoid receptors (DP1, DP2, EP1, EP4, FP, TP) and leaves open IP and EP2 receptors so that their anti-inflammatory properties could be exerted, produced superior inhibitory effects on cytokine release from human macrophages compared to cyclooxygenase (COX) inhibitors. This favorable activity profile translated into animal studies, with AGN 211377 exceeding the level of inhibition afforded by COX inhibition. AGN 211377 was not, however, a practical drug candidate, having poor bioavailability and cost of goods concerns. Compound 1 (designated AGN 225660) represents a second-generation compound with an entirely different "druggable" core structure. Such a dramatic change in chemical scaffold created uncertainty with respect to matching the effects of AGN 211377. AGN 225660 inhibited RANTES, IL-8, and MCP-1 secretion by at least 50%, from TNFα activated human macrophages. Although AGN 225660 reduced TNFα-evoked MCP-1 release from human monocyte-derived macrophages, it increased LPS-induced MCP-1 secretion (up to 2-fold) from human monocyte-derived dendritic cells. However, AGN 225660 inhibited the release of IL12p 70 and IL-23 from human monocyte-derived dendritic cells stimulated by LPS by more than 70%. This effect of AGN 225660 was reproduced in part by the prototype compound AGN 211377 and a combination of selective DP1, EP1, EP4, FP, and TP antagonists. These findings suggest important effects on T cell skewing and disease modification by this class of therapeutic agents. AGN 225660 exhibited good ocular bioavailability and was active in reducing ocular inflammation associated with phacoemulsification surgery, LPS, and arachidonic acid induced uveitis.

10.
J Ocul Pharmacol Ther ; 35(2): 98-105, 2019 03.
Article in English | MEDLINE | ID: mdl-30547709

ABSTRACT

PURPOSE: The purpose of these studies was (1) to investigate the ability of human M1 phenotype macrophages to secrete vascular endothelial growth factor (VEGF) and the influence of prostacyclin receptor (IP) stimulation (2) to evaluate the contribution of the proangiogenic prostanoid prostacyclin to experimental choroidal neovascularization Methods: Human macrophages derived from primary blood mononuclear cells were functionally biased toward the M1 phenotype by using tumor necrosis factor α (TNFα). Experimental choroidal neovascularization was produced by laser photocoagulation. Antagonist drugs RO-3244794 (IP antagonist) and GW 627368 (EP4 antagonist) were administered according to an optimal dosing regimen that was predetermined by bioavailability studies. RESULTS: IP receptor stimulation had diametrically opposed effects on VEGF release compared with reported data on cytokine/chemokine secretion from human macrophages. For example, the IP agonist cicaprost stimulated VEGF secretion although it inhibits monocyte chemoattractant protein-1 (MCP-1) secretion: both would favor a proangiogenic effect. The IP receptor antagonist RO-3244794 produced an ∼20% statistically significant reduction in the neovascularized lesion area in the choroidal neovascularization model, which was a similar level to that produced by the EP4 antagonist GW 627368. Combining the 2 drugs produced a statistically significant reduction in neovascularization but only of slightly greater magnitude than that obtained with each antagonist administered alone. CONCLUSIONS: IP receptor stimulation potently and highly efficaciously promoted VEGF release from human M1 macrophages, indicating a possible contribution of the M1 macrophage subtype to VEGF-induced choroidal neovascularization. Studies in living animals suggest that prostacyclin and its target IP receptor contribute to choroidal neovascularization, although to a more modest extent than might have been expected.


Subject(s)
Antihypertensive Agents/pharmacology , Choroidal Neovascularization/drug therapy , Epoprostenol/pharmacology , Macrophages/drug effects , Ophthalmic Solutions/pharmacology , Animals , Cells, Cultured , Chemokine CCL2/analysis , Chemokine CCL2/deficiency , Chemokine CCL2/metabolism , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Humans , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factors/analysis , Vascular Endothelial Growth Factors/metabolism
11.
J Ocul Pharmacol Ther ; 35(9): 469-474, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31329508

ABSTRACT

For >2 decades, EP2 agonists have been the subject of antiglaucoma research and development by scientists in industry and academia around the world. The road has led to the recent approval of the first drug of this class. This article reviews the development of EP2 agonists from conception to clinical approval, discussing pharmacology, structure, biodistribution, therapeutics, and drug delivery. An extensive list of source references is provided for the reader's benefit.


Subject(s)
Antihypertensive Agents/pharmacology , Glaucoma/drug therapy , Receptors, Prostaglandin E, EP2 Subtype/agonists , Animals , Antihypertensive Agents/chemistry , Drug Delivery Systems , Glaucoma/metabolism , Humans , Receptors, Prostaglandin E, EP2 Subtype/metabolism
12.
J Ocul Pharmacol Ther ; 35(5): 265-277, 2019 06.
Article in English | MEDLINE | ID: mdl-31025909

ABSTRACT

Purpose: Two features define the future of glaucoma therapeutics: (1) greatly improved ocular hypotensive efficacy and (2) a delivery method that improves patient convenience and compliance. A highly efficacious and extraordinarily long-acting ocular hypotensive agent PGN 9856-isopropyl ester represents a potential next-generation anti-glaucoma drug. A new periorbital drug delivery route was also investigated. Methods: PGN 9856-isopropyl ester pharmacology was determined by employing human cells, including prostanoid receptor transfectants, and FLIPr or cellular dielectric spectroscopy technology. Intraocular pressure (IOP) was measured in conscious cynomolgus monkeys trained to accept pneumatonometry when under gentle restraint. For periorbital application, the compound was applied radially using a roller-ball device connected to a cylindrical reservoir. Pharmacokinetic data were obtained using LC/MS/MS instrumentation. Results: Single doses of PGN 9856-isopropyl ester, administered over a 0.001%-0.01% dose range, produced profound decreases in monkey IOP that persisted for at least 5 days, which was long after the drug was detectable in ocular tissues. It was not uncommon for a single eye drop to reduce IOP to the level of 4-7 mm Hg. Drug application to the periorbital dermis of ocular normotensive monkeys produced a similarly profound reduction in IOP, which was well maintained. Conclusions: PGN 9856-isopropyl ester appears to possess efficacy and duration of action properties unmatched by currently prescribed anti-glaucoma agents and by those currently undergoing clinical evaluation. In addition, application to the periorbital skin using a roller-ball device offers a more convenient method of ophthalmic drug delivery than eye drops and is noninvasive, unlike other "dropless" technologies.


Subject(s)
Acetates/therapeutic use , Biphenyl Compounds/therapeutic use , Drug Delivery Systems , Esters/therapeutic use , Glaucoma/drug therapy , Ophthalmic Solutions/therapeutic use , Acetates/administration & dosage , Acetates/chemistry , Administration, Topical , Animals , Aqueous Humor/chemistry , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/chemistry , Cornea/chemistry , Esters/administration & dosage , Esters/chemistry , Female , Humans , Macaca fascicularis , Male , Ophthalmic Solutions/administration & dosage , Ophthalmic Solutions/chemistry , Optical Imaging , Time Factors
13.
ACS Chem Neurosci ; 10(1): 424-437, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30226747

ABSTRACT

The discovery of extended catalytic versatilities is of great importance in both the chemistry and biotechnology fields. Fatty acid amide hydrolase (FAAH) belongs to the amidase signature superfamily and is a major endocannabinoid inactivating enzyme using an atypical catalytic mechanism involving hydrolysis of amide and occasionally ester bonds. FAAH inhibitors are efficacious in experimental models of neuropathic pain, inflammation, and anxiety, among others. We report a new multitarget drug, AGN220653, containing a carboxyamide-4-oxazole moiety and endowed with efficacious analgesic and anti-inflammatory activities, which are partly due to its capability of achieving inhibition of FAAH, and subsequently increasing the tissue concentrations of the endocannabinoid anandamide. This inhibitor behaves as a noncompetitive, slowly reversible inhibitor. Autoradiography of purified FAAH incubated with AGN220653, opportunely radiolabeled, indicated covalent binding followed by fragmentation of the molecule. Molecular docking suggested a possible nucleophilic attack by FAAH-Ser241 on the carbonyl group of the carboxyamide-4-oxazole moiety, resulting in the cleavage of the C-C bond between the oxazole and the carboxyamide moieties, instead of either of the two available amide bonds. MRM-MS analyses only detected the Ser241-assisted formation of the carbamate intermediate, thus confirming the cleavage of the aforementioned C-C bond. Quantum mechanics calculations were fully consistent with this mechanism. The study exemplifies how FAAH structural features and mechanism of action may override the binding and reactivity propensities of substrates. This unpredicted mechanism could pave the way to the future development of a completely new class of amidase inhibitors, of potential use against pain, inflammation, and mood disorders.


Subject(s)
Amidohydrolases/metabolism , Analgesics/chemistry , Analgesics/metabolism , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cinnamates/chemistry , Cinnamates/metabolism , Drug Delivery Systems/methods , Analgesics/administration & dosage , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Carbon/chemistry , Carbon/metabolism , Catalysis , Cinnamates/administration & dosage , Mice , Molecular Docking Simulation/methods , Neuralgia/drug therapy , Neuralgia/metabolism , Rats
14.
Invest Ophthalmol Vis Sci ; 48(9): 4107-15, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17724194

ABSTRACT

PURPOSE: Despite structural similarity with prostaglandin F(2 alpha), the ocular hypotensive agent bimatoprost (Lumigan; Allergan, Inc., Irvine, CA) shows unique pharmacology in vitro and functional activity in vivo. Unfortunately, the precise mechanisms that underlie bimatoprost's distinctive impact on aqueous humor dynamics are unclear. The purpose of the present study was to investigate the effects of bimatoprost and a novel prostamide-selective antagonist AGN 211334 on human conventional drainage. METHODS: Two model systems were used to test the consequences of bimatoprost and/or AGN 211334 treatment on conventional drainage. Human anterior segments in organ culture were perfused at a constant flow rate of 2.5 microL/min while pressure was recorded continuously. After stable baseline facilities were established, segments were treated with drug(s), and pressure was monitored for an additional 3 days. In parallel, the drugs' effects on hydraulic conductivity of human trabecular meshwork (TM) cell monolayers were evaluated. Pharmacological properties of AGN 211334 were characterized in isolated feline iris preparations in organ culture and heterologously expressed G-protein-coupled receptors were examined in vitro. RESULTS: Bimatoprost increased outflow facility by an average of 40% +/- 10% within 48 hours of treatment (n = 10, P < 0.001). Preincubation or coincubation with AGN 211334 significantly blunted bimatoprost's effects by 95% or 43%, respectively. Similar results were obtained in cell culture experiments in which bimatoprost increased hydraulic conductivity of TM cell monolayers by 78% +/- 25%. Pretreatment with AGN 211334 completely blocked bimatoprost's effects, while coincubation decreased its effects on average by 74%. In both models, AGN 211334 alone significantly decreased fluid flux across trabecular tissues and cells. CONCLUSIONS: The findings indicate that bimatoprost interacts with a prostamide receptor in the trabecular meshwork to increase outflow facility.


Subject(s)
Amides/pharmacology , Antihypertensive Agents/pharmacology , Cloprostenol/analogs & derivatives , Lipids/pharmacology , Trabecular Meshwork/drug effects , Aged , Aged, 80 and over , Amides/antagonists & inhibitors , Animals , Aqueous Humor/metabolism , Bimatoprost , Calcium/metabolism , Calcium Signaling/physiology , Cats , Cell Culture Techniques , Cloprostenol/antagonists & inhibitors , Cloprostenol/pharmacology , Dinoprost/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Female , Humans , Intraocular Pressure/physiology , Iris/drug effects , Lipids/antagonists & inhibitors , Male , Middle Aged , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Organ Culture Techniques , Oxazoles/pharmacology , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/metabolism , Recombinant Proteins , Trabecular Meshwork/metabolism
15.
J Ocul Pharmacol Ther ; 33(3): 186-192, 2017 04.
Article in English | MEDLINE | ID: mdl-28072560

ABSTRACT

PURPOSE: To investigate the therapeutic potential of a prostacyclin (IP) receptor agonist for ocular inflammation and the effect on immune cells. METHODS: The anti-inflammatory activities of cicaprost were determined in primary human monocyte-derived macrophages and human monocyte-derived dendritic cells (MoDC), as well as a lipopolysaccharides (LPS)-induced rat uveitis model. Multiple cytokine release was measured by utilizing Luminex Technology. Prostacyclin (IP) Receptor expression was detected by reverse transcription-polymerase chain receptor. Leukocyte infiltration and protein exudation in the rat uveitis model were measured using a hemocytometer and protein concentration by a NanoDrop instrument. RESULTS: Cicapost, an IP receptor agonist, potently inhibits proinflammatory chemokines/cytokine production not only from LPS- or TNFα (tumor necrosis factor-alpha)-induced primary human monocyte-derived macrophages, but also from LPS-stimulated MoDC. While constitutively expressed in macrophages, the IP receptor was inducible by LPS stimulation in MoDCs. In a LPS-induced rat uveitis model, cicaprost efficaciously prevents ocular inflammatory cell and protein leakage, as well as inflammatory cytokine release. CONCLUSION: The IP receptor agonist cicaprost is a potent anti-inflammatory agent, implicating that the tightly controlled PGI2/IP signaling pathway is important in regulating inflammation. This response could be harnessed in ocular inflammatory disease where steroids are currently the standard of care.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dendritic Cells/drug effects , Disease Models, Animal , Epoprostenol/analogs & derivatives , Macrophages/drug effects , Uveitis/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Dendritic Cells/metabolism , Epoprostenol/administration & dosage , Epoprostenol/pharmacology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/antagonists & inhibitors , Macrophages/metabolism , Male , Rats, Inbred Lew , Receptors, Epoprostenol/agonists , Receptors, Epoprostenol/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Uveitis/chemically induced
16.
J Colloid Interface Sci ; 450: 417-423, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25863224

ABSTRACT

HYPOTHESIS: Nanoparticle adsorption at the oil-water interface in an unstable, coalescing emulsion leads to cluster formation. EXPERIMENTS: Stable suspensions of clusters are prepared using a facile, two-step procedure involving few reagents and neither thiolated compounds nor chlorinated solvents. First, colloidal gold nanoparticles are assembled at the aqueous-hexanol interface in an emulsion that rapidly coalesces and spontaneously deposits a film on the interior surface of the glass container. The film is dissolved in ethanol with sonication to disperse the clusters. The film and clusters are characterized by transmission electron and atomic force microscopies as well as ultraviolet-visible spectrometry. FINDINGS: Clusters are observed to contain as few as 8 to as many as 24 Au nanoparticles. The clusters are anisotropic and can also be formed from larger nanoparticles. Hydrophobic and hydrophilic interactions are implicated in the formation of these clusters within the interfacial tension gradients of a coalescing emulsion. The clusters can be re-suspended in ethanol and water, maximizing the utility of these clusters with an extinction band in the near-Infrared region of the electromagnetic spectrum.

17.
Invest Ophthalmol Vis Sci ; 54(7): 4782-90, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23766471

ABSTRACT

PURPOSE: The goal of this study was to functionally compare prostaglandin E2 (PGE2)-sensitive receptors in human primary cells involved in conventional outflow. METHODS: The expression profile of prostaglandin (PG) receptors in primary cultures of human trabecular meshwork (TM) and Schlemm's canal (SC) cells were determined by quantitative-PCR. The functional activities of endogenous PGE2-sensitive receptors were evaluated using subtype-selective agonists and antagonists with cell impedance technology. RESULTS: Agonist-sensitive EP1, EP2, and EP4 receptors were present in TM cells, all increasing cell stiffness (or contractility) in a dose-dependent manner. Rank order of efficacy (Emax) for agonists in TM cells were EP1 greater than EP2 greater than EP4 with EC50 1.1 µM, 0.56 µM, and 0.1 µM, respectively, and no functional EP3 receptors were found. Of the four EP receptor subtypes active in SC cells, EP1 and EP3 receptor activation increased cell stiffness, while EP2 and EP4 agonists dose-dependently decreased cell stiffness 47% and 23% with EC50 values of 170 nM and 69 nM, respectively. Consistent with these observations, the Rho kinase inhibitor Y-27632 decreased cell impedance (stiffness) of TM and SC cells (∼60%), while Rho GTPase activator thrombin caused cell impedance to increase in both cell types (168%-190%). CONCLUSIONS: Cell impedance positively correlates with cellular stiffness/contractility. Because EP2/4 receptors caused decreased cell stiffness in SC, but not in TM cells, both receptors appear to mediate IOP lowering via changes in SC cell stiffness in the conventional outflow pathway.


Subject(s)
Elasticity/physiology , Receptors, Prostaglandin/metabolism , Trabecular Meshwork/physiology , Amides/pharmacology , Cells, Cultured , Dinoprostone/metabolism , Dinoprostone/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Polymerase Chain Reaction , Pyridines/pharmacology , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/drug effects , Trabecular Meshwork/drug effects
18.
Invest Ophthalmol Vis Sci ; 51(10): 5176-81, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20435598

ABSTRACT

PURPOSE: Bimatoprost is a widely used ocular hypotensive agent to treat glaucoma. It lowers intraocular pressure in humans by increasing both pressure-independent (uveoscleral) and pressure-dependent (conventional) aqueous humor outflow. The present study specifically examines bimatoprost effects on the cells that populate human outflow tissues. METHODS: The authors tested for prostamide receptor activation in primary cultures of human trabecular meshwork (TM), Schlemm's canal (SC), and ciliary smooth muscle (CSM) cells using cellular dielectric spectroscopy (CDS). RESULTS: The authors observed that bimatoprost produced an immediate and concentration-dependent increase in cell monolayer impedance for TM, SC, and CSM cells with EC(50) values of 4.3, 1.2, and 1.7 nM, respectively; corresponding to decreased cell contractility. Notably, in TM, SC, and CSM cells, bimatoprost was approximately equipotent to the selective FP receptor agonists fluprostenol and 17-phenyl PGF(2α). Bimatoprost effects were insensitive to cholera toxin and pertussis toxin but were abolished by phorbol 12-myristate 13-acetate pretreatment, suggesting Gq-involvement in cell signaling. The effects of bimatoprost on TM and SC cells were inhibited by the prostamide receptor antagonist AGN211334, with IC(50) values of 1.2 and 3.3 µM, respectively. Interestingly, AGN211334 behaved as an apparent inverse agonist in CDS assays involving TM cells but as a neutral prostamide antagonist with SC cells. CONCLUSIONS: Taken together, results suggest that bimatoprost specifically activates receptors in both cell types of the human conventional outflow pathway to modify intraocular pressure. However, only TM cell monolayers appear to have autocrine, or agonist-independent, receptor signaling that is sensitive to a prostamide receptor antagonist.


Subject(s)
Amides/pharmacology , Antihypertensive Agents/pharmacology , Aqueous Humor/metabolism , Ciliary Body/drug effects , Cloprostenol/analogs & derivatives , Cornea/drug effects , Muscle, Smooth/drug effects , Trabecular Meshwork/drug effects , Actins/metabolism , Adult , Aged , Amides/antagonists & inhibitors , Antihypertensive Agents/antagonists & inhibitors , Bimatoprost , Cells, Cultured , Ciliary Body/metabolism , Cloprostenol/antagonists & inhibitors , Cloprostenol/pharmacology , Cornea/metabolism , Dose-Response Relationship, Drug , Electric Impedance , Humans , Infant , Intraocular Pressure/drug effects , Middle Aged , Muscle, Smooth/metabolism , Receptors, Prostaglandin/metabolism , Tissue Donors , Trabecular Meshwork/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL