Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Orthop Res ; 39(9): 2048-2061, 2021 09.
Article in English | MEDLINE | ID: mdl-33104243

ABSTRACT

The Wnt pathway is upregulated in tendinopathy, affecting inflammation and tenocyte differentiation. Given its potential role in tendinopathy, this signaling pathway may be a relevant target for treatment. The current study examined the therapeutic potential of SM04755, a topical, small-molecule Wnt pathway inhibitor, for the treatment of tendinopathy using in vitro assays and animal models. In vitro, SM04755 decreased Wnt pathway activity, induced tenocyte differentiation, and inhibited catabolic enzymes and pro-inflammatory cytokines in human mesenchymal stem cells, rat tendon-derived stem cells, and human peripheral blood mononuclear cells. Evaluation of the mechanism of action of SM04755 by biochemical profiling and computational modeling identified CDC-like kinase 2 (CLK2) and dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) as molecular targets. CLK and DYRK1A inhibition by siRNA knockdown or pharmacological inhibition induced tenocyte differentiation and reduced tenocyte catabolism. In vivo, topically applied SM04755 showed therapeutically relevant exposure in tendons with low systemic exposure and no detectable toxicity in rats. Moreover, SM04755 showed reduced tendon inflammation and evidence of tendon regeneration, decreased pain, and improved weight-bearing function in rat collagenase-induced tendinopathy models compared with vehicle control. Together, these data demonstrate that CLK2 and DYRK1A inhibition by SM04755 resulted in Wnt pathway inhibition, enhanced tenocyte differentiation and protection, and reduced inflammation. SM04755 has the potential to benefit symptoms and modify disease processes in tendinopathy.


Subject(s)
Tendinopathy , Wnt Signaling Pathway , Animals , Inflammation , Leukocytes, Mononuclear , Rats , Tendinopathy/drug therapy , Tendinopathy/metabolism , Tendons
2.
Cancer Lett ; 473: 186-197, 2020 03 31.
Article in English | MEDLINE | ID: mdl-31560935

ABSTRACT

The Wnt/ß-catenin signaling pathway is aberrantly activated in colorectal (CRC) and many other cancers, and novel strategies for effectively targeting it may be needed due to its complexity. In this report, SM08502, a novel small molecule in clinical development for the treatment of solid tumors, was shown to reduce Wnt pathway signaling and gene expression through potent inhibition of CDC-like kinase (CLK) activity. SM08502 inhibited serine and arginine rich splicing factor (SRSF) phosphorylation and disrupted spliceosome activity, which was associated with inhibition of Wnt pathway-related gene and protein expression. Additionally, SM08502 induced the generation of splicing variants of Wnt pathway genes, suggesting that its mechanism for inhibition of gene expression includes effects on alternative splicing. Orally administered SM08502 significantly inhibited growth of gastrointestinal tumors and decreased SRSF phosphorylation and Wnt pathway gene expression in xenograft mouse models. These data implicate CLKs in the regulation of Wnt signaling and represent a novel strategy for inhibiting Wnt pathway gene expression in cancers. SM08502 is a first-in-class CLK inhibitor being investigated in a Phase 1 clinical trial for subjects with advanced solid tumors (NCT03355066).


Subject(s)
Colorectal Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Serine-Arginine Splicing Factors/metabolism , Stomach Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Alternative Splicing/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Inhibitory Concentration 50 , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Rats , Stomach Neoplasms/pathology , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
3.
Aging Cell ; 18(5): e13000, 2019 10.
Article in English | MEDLINE | ID: mdl-31267651

ABSTRACT

Dual-specificity tyrosine phosphorylation-regulated kinase-1A (DYRK1A) is known to phosphorylate the microtubule-associated tau protein. Overexpression is correlated with tau hyperphosphorylation and neurofibrillary tangle (NFT) formation in Alzheimer's disease (AD). This study assessed the potential of SM07883, an oral DYRK1A inhibitor, to inhibit tau hyperphosphorylation, aggregation, NFT formation, and associated phenotypes in mouse models. Exploratory neuroinflammatory effects were also studied. SM07883 specificity was tested in a kinase panel screen and showed potent inhibition of DYRK1A (IC50  = 1.6 nM) and GSK-3ß (IC50  = 10.8 nM) kinase activity. Tau phosphorylation measured in cell-based assays showed a reduction in phosphorylation of multiple tau epitopes, especially the threonine 212 site (EC50  = 16 nM). SM07883 showed good oral bioavailability in multiple species and demonstrated a dose-dependent reduction of transient hypothermia-induced phosphorylated tau in the brains of wild-type mice compared to vehicle (47%, p < 0.001). Long-term efficacy assessed in aged JNPL3 mice overexpressing the P301L human tau mutation (3 mg/kg, QD, for 3 months) exhibited significant reductions in tau hyperphosphorylation, oligomeric and aggregated tau, and tau-positive inclusions compared to vehicle in brainstem and spinal cord samples. Reduced gliosis compared to vehicle was further confirmed by ELISA. SM07883 was well tolerated with improved general health, weight gain, and functional improvement in a wire-hang test compared to vehicle-treated mice (p = 0.048). SM07883, a potent, orally bioavailable, brain-penetrant DYRK1A inhibitor, significantly reduced effects of pathological tau overexpression and neuroinflammation, while functional endpoints were improved compared to vehicle in animal models. This small molecule has potential as a treatment for AD.


Subject(s)
Alzheimer Disease/drug therapy , Isoquinolines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , tau Proteins/metabolism , Administration, Oral , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Isoquinolines/administration & dosage , Isoquinolines/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Structure , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , tau Proteins/genetics , tau Proteins/toxicity , Dyrk Kinases
4.
Audiol Neurootol ; 21(1): 22-9, 2016.
Article in English | MEDLINE | ID: mdl-26789647

ABSTRACT

The otoprotective effects of OTO-104 were investigated following both acute and chronic administration of cisplatin. The acute administration of cisplatin to guinea pigs resulted in profound hearing loss (70-80 dB SPL) across all frequencies tested. A single intratympanic injection of 6% OTO-104, but not of lower doses, almost completely protected against cisplatin ototoxicity. In contrast, a dexamethasone solution administered under the same experimental conditions offered no otoprotection. OTO-104 was also very effective in protecting against the progressive hearing loss observed with the chronic administration of cisplatin (3 injections at a weekly interval). The otoprotection was found to be dependent upon the activation of dexamethasone-dependent classical nuclear receptor pathways.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Dexamethasone/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Glucocorticoids/pharmacology , Hearing Loss, Sensorineural/chemically induced , Hearing/drug effects , Animals , Dexamethasone/administration & dosage , Dexamethasone/analogs & derivatives , Disease Models, Animal , Female , Glucocorticoids/administration & dosage , Guinea Pigs , Hearing Loss, Sensorineural/prevention & control , Hydrogel, Polyethylene Glycol Dimethacrylate , Injection, Intratympanic , Poloxamer
5.
Audiol Neurootol ; 21(1): 12-21, 2016.
Article in English | MEDLINE | ID: mdl-26655654

ABSTRACT

The otoprotective effects of OTO-104 were investigated both prior to and following acute acoustic trauma. Guinea pigs received a single intratympanic injection of OTO-104 and were assessed in a model of acute acoustic trauma. Doses of at least 2.0% OTO-104 offered significant protection against hearing loss induced by noise exposure when administered 1 day prior to trauma and up to 3 days thereafter. Otoprotection remained effective even with higher degrees of trauma. In contrast, the administration of a dexamethasone sodium phosphate solution did not protect against noise-induced hearing loss. Activation of the classical nuclear glucocorticoid and mineralocorticoid receptor pathways was required for otoprotection by OTO-104. The sustained exposure properties of OTO-104 were also superior to a steroid solution.


Subject(s)
Dexamethasone/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Glucocorticoids/pharmacology , Hearing Loss, Noise-Induced/prevention & control , Hearing/drug effects , Animals , Dexamethasone/administration & dosage , Dexamethasone/analogs & derivatives , Disease Models, Animal , Female , Glucocorticoids/administration & dosage , Guinea Pigs , Hydrogel, Polyethylene Glycol Dimethacrylate , Injection, Intratympanic , Poloxamer
6.
Otol Neurotol ; 35(3): 459-69, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24518407

ABSTRACT

HYPOTHESIS: OTO-201 can provide sustained release to the middle ear and effectively treat otitis media, when compared with FDA-approved ciprofloxacin otic drop formulations. BACKGROUND: There is an unmet medical need for antibiotic therapy that can provide a full course of treatment from a single administration by an otolaryngologist at the time of tympanostomy tube placement, obviating the need for twice daily multiday treatment with short-acting otic drops. METHODS: Studies in guinea pigs and chinchillas were conducted. OTO-201 was administered as a single intratympanic injection and compared with the twice daily multi-day treatment with Ciprodex or Cetraxal otic drops. RESULTS: OTO-201 demonstrated sustained release of ciprofloxacin in the middle ear compartment for days to approximately 2 weeks depending on the dose. The substantial C(max) values and steady drug exposure yielded by OTO-201 were in contrast to the pulsatile short lasting exposure seen with Ciprodex and Cetraxal. OTO-201 was also effective in a preclinical chinchilla model of Streptococcus pneumoniae-induced otitis media. The degree of cure was comparable to that afforded by Ciprodex and Cetraxal. There was no evidence of middle or inner ear pathology in guinea pigs treated with OTO-201, unlike Ciprodex and Cetraxal, which both demonstrated mild cochlear ototoxicity. No adverse effects of the poloxamer 407 vehicle were noted. CONCLUSION: Intratympanic injection of OTO-201 constitutes an attractive treatment option to twice daily multiday dosing with ciprofloxacin ear drops for the treatment of otitis media, as evidenced by superior middle ear drug exposure, efficacy in an acute otitis media model, safety of administration, and convenience of a single dose regimen.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Ciprofloxacin/therapeutic use , Delayed-Action Preparations/therapeutic use , Hydrogels/therapeutic use , Otitis Media/drug therapy , Administration, Topical , Animals , Anti-Bacterial Agents/administration & dosage , Chinchilla , Ciprofloxacin/administration & dosage , Delayed-Action Preparations/administration & dosage , Disease Models, Animal , Guinea Pigs , Hydrogels/administration & dosage
7.
Laryngoscope ; 121(2): 385-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21271594

ABSTRACT

OBJECTIVE/HYPOTHESIS: Previous studies revealed that intratympanic administration of the steroid dexamethasone in poloxamer 407 hydrogel, a class of thermoreversible polymers, resulted in significant and durable exposure in the inner ear. Interestingly, varying the concentrations of the poloxamer vehicle and of the steroid impacted the pharmacokinetic profile of dexamethasone in the perilymphatic compartment. Here, the respective contributions of different vehicles (aqueous solution, poloxamer hydrogel) and steroid drugs (dexamethasone, methylprednisolone) were investigated. In particular, various forms of the steroids, discriminated by their aqueous solubility, were compared. STUDY DESIGN: In vitro studies characterized the gelation profile and drug release kinetics of the various formulations. The inner ear pharmacokinetic profile of the different formulations was investigated in guinea pigs. RESULTS: Drugs formulated in poloxamer 407 shared significantly more prolonged exposure than those formulated in aqueous solutions both in vitro and in vivo in the inner ear. Furthermore, drugs with low aqueous solubility yielded increased degree and duration of exposure in the inner ear relative to water-soluble drugs. CONCLUSIONS: The inner ear pharmacokinetic profile of drugs administered intratympanically is not only highly dependent upon the nature of the vehicle but also upon the physicochemical properties of the drug delivered.


Subject(s)
Delayed-Action Preparations/administration & dosage , Ear, Inner/drug effects , Steroids/administration & dosage , Tympanic Membrane , Animals , Dexamethasone/administration & dosage , Dexamethasone/pharmacokinetics , Guinea Pigs , Methylprednisolone/administration & dosage , Methylprednisolone/pharmacokinetics , Poloxamer/administration & dosage , Steroids/pharmacokinetics
8.
Audiol Neurootol ; 16(4): 233-41, 2011.
Article in English | MEDLINE | ID: mdl-20980743

ABSTRACT

Information on inner ear pharmacokinetics is limited in the literature, especially in large animals and in humans. A preliminary study was designed to explore the differences in inner ear exposure between guinea pigs and sheep following a single intratympanic injection of a 2% dexamethasone sodium phosphate solution. In both species, significant levels of dexamethasone were observed in the perilymph within 1 h, and decreasing by 50- to 100-fold within 12 h. Overall, the exposure to dexamethasone in the inner ear was significantly lower in sheep by 17- to 27-fold than in guinea pigs. Systemic and CNS exposure were minimal in both species as indicated by the low drug levels observed in plasma and CSF. Altogether, the preliminary evidence presented herein suggests the sheep as a practical and acceptable animal model to study the inner ear pharmacokinetics of drug candidates in large mammals and its potential towards extrapolation to human exposure.


Subject(s)
Dexamethasone/pharmacokinetics , Tympanic Membrane , Animals , Female , Guinea Pigs , Injections , Perilymph , Sheep
9.
Otol Neurotol ; 32(1): 171-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21099726

ABSTRACT

HYPOTHESIS: To investigate whether OTO-104, a poloxamer-based hydrogel containing micronized dexamethasone for intratympanic delivery, can provide long-lasting inner ear exposure and be well tolerated. METHODS: OTO-104 was administered intratympanically to guinea pigs and sheep, and its pharmacokinetic and toxicity profiles were examined. RESULTS: After a single intratympanic injection of OTO-104 (from 0.6% to 20%, w/w), significant and prolonged exposure to dexamethasone in the inner ear was observed. Increasing the concentration of OTO-104 resulted in higher perilymph drug levels as well as a more prolonged duration of exposure. At the highest dose, therapeutic perilymph levels of dexamethasone could be sustained over 3 months in guinea pigs and more than 1 month in sheep. A toxicologic evaluation was conducted, including assessments of middle and inner ear function and physiology, as well as appraisal of local and systemic toxicity. A small and transient shift in hearing threshold was observed, most probably conductive in nature. No significant histologic changes in middle or inner ear tissues were noted. Although macroscopically mild erythema/inflammation was documented in a subset of guinea pigs treated with 20% OTO-104, the nature and the severity of these changes were not different between the poloxamer vehicle, saline, and 20% OTO-104 groups. No evidence of acute dermal toxicity, delayed hypersensitivity, or systemic adverse effects was found. CONCLUSION: OTO-104 is a novel proprietary therapeutic delivery system that can achieve prolonged, sustained release of dexamethasone within the inner ear fluids. The administration of this clinical candidate formulation via intratympanic injection is expected to be well tolerated both locally and systemically.


Subject(s)
Dexamethasone/administration & dosage , Ear, Inner/chemistry , Hydrogels/administration & dosage , Perilymph/chemistry , Animals , Delayed-Action Preparations , Dexamethasone/analysis , Dexamethasone/pharmacokinetics , Guinea Pigs , Hydrogels/analysis , Hydrogels/pharmacokinetics , Injections , Sheep
10.
Audiol Neurootol ; 14(6): 393-401, 2009.
Article in English | MEDLINE | ID: mdl-19923809

ABSTRACT

The thermo-reversible triblock copolymer poloxamer 407 was investigated as a drug delivery vehicle for micronized dexamethasone into the middle and inner ears of guinea pigs. The study characterized the gelation and in vitro release kinetics of poloxamer formulations. In vivo, the pharmacokinetic profile of formulations containing varying concentrations of poloxamer and dexamethasone was examined following intratympanic administration. Significant drug levels within the perilymph were observed for at least 10 days, while systemic exposure was minimal. The sustained-release kinetics profile could be significantly modulated by varying the concentrations of both poloxamer and dexamethasone. Assessment of auditory function revealed a small transient shift in hearing threshold, most probably of conductive nature, which resolved itself within a week. No significant histological changes of the round window membrane or cochlea could be noted. Poloxamer 407 thus represents an effective and safe delivery system to achieve sustained release of dexamethasone to the inner ear.


Subject(s)
Dexamethasone/administration & dosage , Dexamethasone/pharmacokinetics , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Perilymph/drug effects , Tympanic Membrane/drug effects , Analysis of Variance , Animals , Cochlea/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Routes , Electrophysiology , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Guinea Pigs , Hearing/drug effects , Hearing Tests , Poloxamer/administration & dosage , Poloxamer/pharmacokinetics
11.
J Pharmacol Exp Ther ; 328(3): 758-65, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19056934

ABSTRACT

Phosphatidylinositol 3-kinases (PI3Ks) are key elements in the signaling cascades that lie downstream of many cellular receptors. In particular, PI3K delta and gamma isoforms contribute to inflammatory cell recruitment and subsequent activation. For this reason, in a series of preclinical studies, we tested the potential of a recently developed small-molecule inhibitor of these two isoforms, TG100-115 [3-[2,4-diamino-6-(3-hydroxyphenyl)pteridin-7-yl]phenol], as a form of anti-inflammatory therapy for respiratory diseases such as asthma and chronic obstructive pulmonary disease (COPD). To determine pharmacokinetic profiles, aerosolized formulations of the drug were delivered to mice by a nose-only inhalation route, yielding high pulmonary TG100-115 levels with minimal systemic exposure. Safety assessments were favorable, with no clinical or histological changes noted after 21 days of daily dosing. In a murine asthma model, aerosolized TG100-115 markedly reduced the pulmonary eosinophilia and the concomitant interleukin-13 and mucin accumulation characteristic of this disease. As a functional benefit, interventional dosing schedules of this inhibitor also reduced airway hyper-responsiveness. To model the pulmonary neutrophilia characteristic of COPD, mice were exposed to either intranasal lipopolysaccharide or inhaled smoke. Aerosolized TG100-115 again inhibited these inflammatory patterns, most notably in the smoke model, where interventional therapy overcame the steroid-resistant nature of the pulmonary inflammation. In conclusion, aerosolized TG100-115 displays pharmacokinetic, safety, and biological activity profiles favorable for further development as a therapy for both asthma and COPD. Furthermore, these studies support the hypothesis that PI3K delta and gamma are suitable molecular targets for these diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Phenols/therapeutic use , Pteridines/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Administration, Intranasal , Aerosols , Animals , Anti-Inflammatory Agents/administration & dosage , Bronchial Hyperreactivity/drug therapy , Class Ib Phosphatidylinositol 3-Kinase , Disease Models, Animal , Isoenzymes/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/toxicity , Mice , Neutrophils/drug effects , Neutrophils/physiology , Phosphatidylinositol 3-Kinases/metabolism
12.
J Cell Physiol ; 216(1): 29-37, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18330892

ABSTRACT

Age-related macular degeneration, diabetic retinopathy, and retinal vein occlusions are complicated by neovascularization and macular edema. Multi-targeted kinase inhibitors that inhibit select growth factor receptor tyrosine kinases and/or components of their down-stream signaling cascades (such as Src kinases) are rationale treatment strategies for these disease processes. We describe the discovery and characterization of two such agents. TG100572, which inhibits Src kinases and selected receptor tyrosine kinases, induced apoptosis of proliferating endothelial cells in vitro. Systemic delivery of TG100572 in a murine model of laser-induced choroidal neovascularization (CNV) caused significant suppression of CNV, but with an associated weight loss suggestive of systemic toxicity. To minimize systemic exposure, topical delivery of TG100572 to the cornea was explored, and while substantial levels of TG100572 were achieved in the retina and choroid, superior exposure levels were achieved using TG100801, an inactive prodrug that generates TG100572 by de-esterification. Neither TG100801 nor TG100572 were detectable in plasma following topical delivery of TG100801, and adverse safety signals (such as weight loss) were not observed even with prolonged dosing schedules. Topical TG100801 significantly suppressed laser-induced CNV in mice, and reduced fluorescein leakage from the vasculature and retinal thickening measured by optical coherence tomography in a rat model of retinal vein occlusion. These data suggest that TG100801 may provide a new topically applied treatment approach for ocular neovascularization and retinal edema.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Papilledema/drug therapy , Phenols/therapeutic use , Prodrugs/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triazines/therapeutic use , src-Family Kinases/antagonists & inhibitors , Administration, Topical , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/metabolism , Animals , Cell Line , Choroidal Neovascularization/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Papilledema/pathology , Prodrugs/adverse effects , Prodrugs/metabolism , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/metabolism , Rabbits , Rats , Rats, Long-Evans , Receptor Protein-Tyrosine Kinases/metabolism , Retina/cytology , Retina/metabolism , Retina/pathology , src-Family Kinases/metabolism
13.
J Med Chem ; 51(6): 1546-59, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18311895

ABSTRACT

Age-related macular degeneration (AMD) is one of the leading causes of loss of vision in the industrialized world. Attenuating the VEGF signal in the eye to treat AMD has been validated clinically. A large body of evidence suggests that inhibitors targeting the VEGFr pathway may be effective for the treatment of AMD. Recent studies using Src/YES knockout mice suggest that along with VEGF, Src and YES play a crucial role in vascular leak and might be useful in treating edema associated with AMD. Therefore, we have developed several potent benzotriazine inhibitors designed to target VEGFr2, Src, and YES. One of the most potent compounds is 4-chloro-3-{5-methyl-3-[4-(2-pyrrolidin-1-yl-ethoxy)phenylamino]benzo[1,2,4]triazin-7-yl}phenol ( 5), a dual inhibitor of both VEGFr2 and the Src family (Src and YES) kinases. Several ester analogues of 5 were prepared as prodrugs to improve the concentration of 5 at the back of the eye after topical administration. The thermal stability of these esters was studied, and it was found that benzoyl and substituted benzoyl esters of 5 showed good thermal stability. The hydrolysis rates of these prodrugs were studied to analyze their ability to undergo conversion to 5 in vivo so that appropriate concentrations of 5 are available in the back-of-the-eye tissues. From these studies, we identified 4-chloro-3-(5-methyl-3-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}-1,2,4-benzotriazin-7-yl)phenyl benzoate ( 12), a topically administered prodrug delivered as an eye drop that is readily converted to the active compound 5 in the eye. This topically delivered compound exhibited excellent ocular pharmacokinetics and poor systemic circulation and showed good efficacy in the laser induced choroidal neovascularization model. On the basis of its superior profile, compound 12 was advanced. It is currently in a clinical trial as a first in class, VEGFr2 targeting, topically applied compound for the treatment of AMD.


Subject(s)
Macular Degeneration/drug therapy , Ophthalmic Solutions/therapeutic use , Phenols/therapeutic use , Prodrugs/therapeutic use , Triazines/therapeutic use , Administration, Topical , Animals , Choroidal Neovascularization/drug therapy , Clinical Trials as Topic , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Eye/drug effects , Eye/radiation effects , Lasers , Mice , Mice, Knockout , Models, Molecular , Molecular Structure , Ophthalmic Solutions/chemistry , Ophthalmic Solutions/pharmacokinetics , Phenols/chemistry , Phenols/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Structure-Activity Relationship , Triazines/chemistry , Triazines/pharmacokinetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors
14.
J Control Release ; 95(3): 489-500, 2004 Mar 24.
Article in English | MEDLINE | ID: mdl-15023460

ABSTRACT

Parenteral administration of immunoglobulins (Ig) for prevention or treatment of respiratory diseases achieves only modest concentrations of antibody in the pulmonary interstitial tissue and airways. Aerosols, including spray-dried particles, must overcome two limiting factors in order to be effective vehicles for pulmonary delivery of Ig: (i) Fc receptor (FcR)-mediated scavenging by macrophages and (ii) clearance by the mucociliary system. Ig-incorporated spray-dried lipid microparticles (SDLM), coformulated with or without a biocompatible surfactant (1% w:w) to modulate protein release, were designed and tested for their capability to deliver Ig to the respiratory tract. To determine efficacy, rodents were immunized with SDLM containing antiinfluenza antibody followed by virus challenge and clinical parameters measured. Control of the release kinetics resulted in enhanced delivery of immunoglobulins to the respiratory tract and interstitial tissue with slow translocation into the systemic circulation. As much as 60% of the IgG delivered from nonretentive SDLM could be recovered from the lung interstitial tissue within 1 h after aerosol administration at a dose of 1 mg of Ig/kg of body weight. In addition, nonretentive rather than slow-release particles loaded with antiinfluenza antibody were effective in curbing virus replication with a resulting positive clinical outcome. Thus, controlled release of Ig by manipulating aerosol characteristics and composition allows for a significant increase in the efficiency of pulmonary delivery of antibodies.


Subject(s)
Aerosols/administration & dosage , Drug Carriers/pharmacokinetics , Drug Design , Immunoglobulin G/administration & dosage , Administration, Inhalation , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/pharmacokinetics , Chemistry, Pharmaceutical/methods , Drug Carriers/administration & dosage , Enzyme-Linked Immunosorbent Assay , Female , Fluorocarbons/chemistry , Fluorocarbons/pharmacokinetics , Lung/chemistry , Lung/drug effects , Mice , Mice, Inbred BALB C , Microspheres , Nebulizers and Vaporizers , Rats , Rats, Sprague-Dawley , Technology, Pharmaceutical
15.
Vaccine ; 21(21-22): 2805-12, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12798621

ABSTRACT

Influenza viruses are among the most significant human pathogens, responsible for increased seasonal morbidity and mortality particularly in immunodepressed and chronically ill. Conventional vaccination with non-replicative vaccine is currently performed by injection. In the present study, we explore simple spray-dried lipid formulations containing whole inactivated virus or split-subunit vaccine that allow aerosolization and thus, mucosal vaccination of the pulmonary tract. We show that by using biocompatible excipients already approved for human use, one could engineer microparticles that induce substantial local and systemic immunity subsequent to pulmonary administration. Exposure of the bronchial-associated lymphoid tissue (BALT) to vaccine was more effective than parenteral or nasal administration in triggering specific immunity. Co-formulation of a biocompatible surfactant detergent greatly ameliorated the immune profile of microparticles containing a whole inactivated virus vaccine. In addition, mere formulation of a licensed split-subunit vaccine significantly enhanced its immunogenicity. Together, our data underline a simple strategy to convert conventional parenteral vaccination of currently available non-replicative vaccines against influenza virus, into one that is more effective and practical upon respiratory administration.


Subject(s)
Influenza Vaccines/immunology , Respiratory Mucosa/immunology , Aerosols , Animals , Bronchi/immunology , Female , Influenza Vaccines/administration & dosage , Lymphoid Tissue/immunology , Mice , Mice, Inbred BALB C , Nasal Mucosa/immunology , Orthomyxoviridae Infections/prevention & control , Rats , Rats, Sprague-Dawley , Time Factors , Vaccination
16.
J Clin Invest ; 110(8): 1175-84, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12393853

ABSTRACT

The adaptive immune response is triggered by recognition of T and B cell epitopes and is influenced by "danger" motifs that act via innate immune receptors. This study shows that motifs associated with noncoding RNA are essential features in the immune response reminiscent of viral infection, mediating rapid induction of proinflammatory chemokine expression, recruitment and activation of antigen-presenting cells, modulation of regulatory cytokines, subsequent differentiation of Th1 cells, isotype switching, and stimulation of cross-priming. The heterogeneity of RNA-associated motifs results in differential binding to cellular receptors, and specifically impacts the immune profile. Naturally occurring double-stranded RNA (dsRNA) triggered activation of dendritic cells and enhancement of specific immunity, similar to selected synthetic dsRNA motifs. Based on the ability of specific RNA motifs to block tolerance induction and effectively organize the immune defense during viral infection, we conclude that such RNA species are potent danger motifs. We also demonstrate the feasibility of using selected RNA motifs as adjuvants in the context of novel aerosol carriers for optimizing the immune response to subunit vaccines. In conclusion, RNA-associated motifs produced during viral infection bridge the early response with the late adaptive phase, regulating the activation and differentiation of antigen-specific B and T cells, in addition to a short-term impact on innate immunity.


Subject(s)
RNA, Untranslated/genetics , RNA, Untranslated/immunology , Adaptation, Physiological , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Viral/biosynthesis , Antigens, Viral/administration & dosage , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , RNA, Untranslated/pharmacology , Rats , Rats, Sprague-Dawley , Vaccination , Viral Vaccines/administration & dosage , Virus Diseases/immunology
17.
Pharm Res ; 19(3): 258-64, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11934231

ABSTRACT

PURPOSE: PulmoSphere particles are specifically engineered for delivery by the pulmonary route with a hollow and porous morphology, physical diameters < 5 microm, and low tap densities (circa 0.1 g x cm(-3)). Deposition of PulmoSphere particles in the human respiratory tract delivered by pressurized metered dose inhaler (pMDI) was compared with deposition of a conventional micronized drug pMDI formulation. METHODS: Nine healthy nonsmoking subjects (5 male, 4 female) completed a two-way crossover gamma scintigraphic study, assessing the lung and oropharyngeal depositions of albuterol sulfate, formulated as 99mTc-radiolabeled PulmoSphere particles or micronized particles (Ventolin Evohaler, GlaxoSmithKline, Ltd.) suspended in HFA-134a propellant. RESULTS: Mean (standard deviation) lung deposition, (% ex-valve dose) was doubled for the PulmoSphere formulation compared with Evohaler pMDI (28.5 (11.3) % vs. 14.5 (8.1) %, P < 0.01), whereas oropharyngeal deposition was reduced (42.6 (9.0) % vs. 72.0 (8.0) %, P < 0.01). Both PulmoSphere and Evohaler pMDIs gave uniform deposition patterns within the lungs. CONCLUSIONS: These data provided "proof of concept" in vivo for the PulmoSphere technology as a method of improving targeting of drugs to the lower respiratory tract from pMDIs, and suggested that the PulmoSphere technology may also be suitable for the delivery of systemically acting molecules absorbed via the lung.


Subject(s)
Drug Delivery Systems/methods , Lung/metabolism , Nebulizers and Vaporizers , Adult , Albuterol/administration & dosage , Chemistry, Pharmaceutical , Cross-Over Studies , Drug Delivery Systems/statistics & numerical data , Female , Humans , Hydrocarbons, Fluorinated/administration & dosage , Lung/drug effects , Male , Microspheres , Middle Aged , Nebulizers and Vaporizers/statistics & numerical data , Porosity , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...