Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Ocul Pharmacol Ther ; 38(6): 459-467, 2022.
Article in English | MEDLINE | ID: mdl-35404132

ABSTRACT

Purpose: To compare ocular pharmacokinetics (PK), systemic absorption, and injectability of triamcinolone acetonide (TA) suprachoroidal (SC) and intravitreal (IVT) suspensions. Methods: New Zealand White (NZW) rabbits received a bilateral injection of 4 mg TA injectable suspension, TRI (TRIESENCE ®; Alcon) through either microneedle-based SC or standard IVT injection. Another group of NZW rabbits received a bilateral SC injection (4 mg) of either TRI or a proprietary TA suspension for SC use, CLS-TA (Clearside Biomedical). Blood and ocular tissues were analyzed for TA over 3 months. Separately, injectability of TRI and CLS-TA through a proprietary SC delivery system (SCS Microinjector®; Clearside Biomedical) was compared using microinjector syringe-plunger glide force measurement. Results: Suprachoroidal delivery of TRI, compared with IVT-TRI, resulted in ∼12-fold higher exposure in the retinal pigment epithelium-choroid-sclera, and comparable exposure in the retina. Conversely, SC-TRI, compared to IVT-TRI, resulted in 460-, 34-, and 22-fold lower exposure in the lens, iris-ciliary body, and vitreous humor, and negligible exposure in the aqueous humor. SC injection of either CLS-TA or TRI resulted in comparable and sustained ocular TA levels. Plasma TA levels were generally undetectable in both studies. A significantly greater and more variable glide force was measured for TRI vs. CLS-TA. Conclusions: Suprachoroidal delivery of TA enabled high and durable TA levels targeted to the chorioretina with limited anterior segment exposure. SC delivery of either CLS-TA or TRI resulted in comparable ocular PK profiles with low systemic exposure; however, CLS-TA required lower and less variable glide force than TRI, potentiating more consistent, controlled administration.


Subject(s)
Glucocorticoids , Triamcinolone Acetonide , Animals , Choroid , Rabbits , Retina , Suspensions
2.
J Control Release ; 349: 1045-1051, 2022 09.
Article in English | MEDLINE | ID: mdl-35868358

ABSTRACT

Drug delivery to the suprachoroidal space (SCS®) has become a clinical reality after the 2021 FDA approval of CLS-TA, a triamcinolone acetonide injectable suspension for suprachoroidal use (XIPERE®), administered via a microneedle-based device, the SCS Microinjector®. Suprachoroidal (SC) delivery facilitates targeting, compartmentalization, and durability of small molecule suspensions, thereby potentially addressing some of the efficacy, safety, and treatment burden limitations of current retinal therapies. Herein, the design features of the SCS Microinjector are reviewed, along with the biomechanics of SC drug delivery. Also presented are preclinical evaluations of SC small molecule suspensions from 4 different therapeutic classes (plasma kallikrein inhibitor, receptor tyrosine kinase inhibitor, corticosteroid, complement factor D inhibitor), highlighting their potential for durability, targeted compartmentalization, and acceptable safety profiles following microinjector-based SC delivery. The clinical evaluations of the safety, tolerability and efficacy of SC delivered triamcinolone further supports potential of SC small molecule suspensions as a clinically viable strategy for the treatment of chorioretinal diseases. Also highlighted are current limitations, key pharmacological considerations, and future opportunities to optimize the SC microinjector platform for safe, effective, and potentially long-acting drug delivery for the treatment of chorioretinal disorders.


Subject(s)
Choroid , Triamcinolone Acetonide , Complement Factor D/pharmacology , Plasma Kallikrein/pharmacology , Protein Kinase Inhibitors/pharmacology , Suspensions
3.
Transl Vis Sci Technol ; 10(7): 19, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34128964

ABSTRACT

Purpose: Axitinib, a tyrosine kinase inhibitor, is a potent inhibitor of vascular endothelial growth factor (VEGF) receptors -1, -2 and -3. Suprachoroidal (SC) delivery of axitinib, combined with pan-VEGF inhibition activity of axitinib, has the potential to provide additional benefits compared to the current standard of care with intravitreal anti-VEGF-A agents. This study evaluated the ocular pharmacokinetics and systemic disposition of axitinib after SC administration in rabbits. Methods: Rabbits received axitinib as either a single SC injection (0.03, 0.10, 1.00, or 4.00 mg/eye; n = 4/group) or a single intravitreal injection (1 mg/eye; n = 4/group) in three separate studies. Axitinib concentrations were measured in several ocular compartments and in plasma at predetermined timepoints for up to 91 days. The pharmacokinetics parameters were estimated by noncompartmental analysis. Results: A single SC injection of axitinib suspension (1 mg/eye) resulted in an 11-fold higher mean axitinib exposure in the posterior eye cup, compared with intravitreal injection. Sustained levels of axitinib in the retinal pigment epithelium-choroid-sclera (RCS) and retina were observed throughout the duration of studies after a single SC axitinib injection (0.1 and 4.0 mg/eye), with low exposure in the vitreous humor, aqueous humor, and plasma. Axitinib levels in the RCS were 3 to 5 log orders higher than the reported in vitro (VEGF receptor-2 autophosphorylation inhibition) 50% inhibitory concentration value after 0.1 and 4.0 mg/eye dose levels throughout the 65-day and 91-day studies, respectively. Conclusions: This study demonstrates that SC axitinib suspension has a favorable pharmacokinetics profile with potential as a long-acting therapeutic candidate targeted to affected choroid and retinal pigment epithelium in neovascular age-related macular degeneration. Translational Relevance: Suprachoroidal axitinib suspension has potential to decrease the treatment burden in neovascular age-related macular degeneration, as a long-acting therapeutic candidate, and could yield greater efficacy, as a potent tyrosine kinase pan-VEGF inhibitor, compared with current standard anti-VEGF-A therapies.


Subject(s)
Choroid , Vascular Endothelial Growth Factor A , Animals , Axitinib , Intravitreal Injections , Rabbits , Retina
4.
Pharmaceutics ; 13(2)2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33671815

ABSTRACT

Suprachoroidal drug delivery technology has advanced rapidly and emerged as a promising administration route for a variety of therapeutic candidates, in order to target multiple ocular diseases, ranging from neovascular age-related macular degeneration to choroidal melanoma. This review summarizes the latest preclinical and clinical progress in suprachoroidal delivery of therapeutic agents, including small molecule suspensions, polymeric entrapped small molecules, gene therapy (viral and nonviral nanoparticles), viral nanoparticle conjugates (VNCs), and cell therapy. Formulation customization is critical in achieving favorable pharmacokinetics, and sustained drug release profiles have been repeatedly observed for multiple small molecule suspensions and polymeric formulations. Novel therapeutic agents such as viral and nonviral gene therapy, as well as VNCs, have demonstrated promise in animal studies. Several of these suprachoroidally-administered therapies have been assessed in clinical trials, including small molecule suspensions of triamcinolone acetonide and axitinib, viral vector RGX-314 for gene therapy, and VNC AU-011. With continued drug delivery research and optimization, coupled with customized drug formulations, suprachoroidal drug delivery may address large unmet therapeutic needs in ophthalmology, targeting affected tissues with novel therapies for efficacy benefits, compartmentalizing therapies away from unaffected tissues for safety benefits, and achieving durability to relieve the treatment burden noted with current agents.

5.
AAPS PharmSciTech ; 11(4): 1526-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20976577

ABSTRACT

Injectable lipid emulsions, for decades, have been clinically used as an energy source for hospitalized patients by providing essential fatty acids and vitamins. Recent interest in utilizing lipid emulsions for delivering lipid soluble therapeutic agents, intravenously, has been continuously growing due to the biocompatible nature of the lipid-based delivery systems. Advancements in the area of novel lipids (olive oil and fish oil) have opened a new area for future clinical application of lipid-based injectable delivery systems that may provide a better safety profile over traditionally used long- and medium-chain triglycerides to critically ill patients. Formulation components and process parameters play critical role in the success of lipid injectable emulsions as drug delivery vehicles and hence need to be well integrated in the formulation development strategies. Physico-chemical properties of active therapeutic agents significantly impact pharmacokinetics and tissue disposition following intravenous administration of drug-containing lipid emulsion and hence need special attention while selecting such delivery vehicles. In summary, this review provides a broad overview of recent advancements in the field of novel lipids, opportunities for intravenous drug delivery, and challenges associated with injectable lipid emulsions.


Subject(s)
Fat Emulsions, Intravenous/chemistry , Fatty Acids, Essential/chemistry , Parenteral Nutrition , Triglycerides/chemistry , Critical Illness , Drug Compounding , Drug Delivery Systems , Fat Emulsions, Intravenous/metabolism , Fat Emulsions, Intravenous/pharmacokinetics , Fatty Acids, Essential/metabolism , Fatty Acids, Essential/pharmacokinetics , Humans , Infusions, Intravenous , Pharmaceutical Vehicles/chemistry , Solubility , Triglycerides/metabolism , Triglycerides/pharmacokinetics
6.
Expert Opin Investig Drugs ; 29(9): 935-945, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32657172

ABSTRACT

INTRODUCTION: Integrins are a family of multi-functional cell-adhesion molecules, heterodimeric receptors that connect extracellular matrix (ECM) to actin cytoskeleton in the cell cortex, thus regulating cellular adhesion, migration, proliferation, invasion, survival, and apoptosis. Consequently, integrins play a role in inflammation, angiogenesis and fibrosis. AREAS COVERED: This review examines individual anti-integrin agents in terms of their chemical nature, route of administration, and anti-integrin action. It also provides a summary of preclinical and clinical studies. Current clinical candidates include risuteganib, THR-687, and SF-0166, which have shown promise in treating diabetic macular edema (DME) and/or age-related macular degeneration (AMD) in early clinical studies. Preclinical candidates include SB-267268, AXT-107, JNJ-26076713, Cilengitide and Lebecetin, which exhibit a decrease in retinal permeability, angiogenesis and/or choroidal neovascularization (CNV). EXPERT OPINION: Anti-integrin therapies show potential in treating retinal diseases. Anti-integrin agents tackle the multi-factorial nature of diabetic retinopathy (DR) and AMD and show promise as injectable and topical agents in preclinical and early clinical studies. Integrin inhibition has potential to serve as primary therapy, adjunctive therapy to anti-vascular endothelial growth factor agents, or secondary therapy in refractory cases.


Subject(s)
Choroidal Neovascularization/drug therapy , Integrins/antagonists & inhibitors , Retinal Diseases/drug therapy , Animals , Choroidal Neovascularization/physiopathology , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/physiopathology , Drug Development , Humans , Macular Degeneration/drug therapy , Macular Degeneration/physiopathology , Macular Edema/drug therapy , Macular Edema/physiopathology , Retinal Diseases/physiopathology
7.
J Ocul Pharmacol Ther ; 36(6): 384-392, 2020.
Article in English | MEDLINE | ID: mdl-32255727

ABSTRACT

Retinal gene therapy is a rapidly growing field with numerous clinical trials underway, and route of delivery is a critical contributor to its success. Subretinal administration, which involves pars plana vitrectomy in the operating room, offers targeted delivery to retinal-pigment epithelium cells and photoreceptors. Due to the immune-privileged nature of the subretinal space, the risk of an immune reaction against viral capsid antigens is minimized, an advantage of subretinal administration in patients with preexisting neutralizing antibodies. Intravitreal administration, with fewer procedure-related complications, is challenged by potential immune response and incomplete vector penetration through the internal limiting membrane. However, novel vectors, optimized by "directed evolution" may address these issues. Nonsurgical in-office suprachoroidal gene delivery offers the potential for greater surface-area coverage of the posterior segment compared to focal subretinal injection, and is not hindered by the internal limiting membrane. However, the vector must pass through multiple layers to reach the targeted retinal layers, and there is a risk of immune response. This review highlights recent developments, challenges, and future opportunities associated with viral and nonviral suprachoroidal gene delivery for the treatment of chorioretinal diseases. While ocular tolerability and short-term effectiveness of suprachoroidal gene delivery have been demonstrated in preclinical models, durability of gene expression, long-term safety, potential systemic exposure, and effective delivery to the macula require further exploration. Although the safety and efficacy of suprachoroidal gene delivery are yet to be proven in clinical trials, further optimization could facilitate nonsurgical in-office suprachoroidal gene therapy.


Subject(s)
Choroid Diseases/therapy , Choroidal Effusions/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Retinal Diseases/therapy , Animals , Capsid Proteins/immunology , Capsid Proteins/therapeutic use , Choroidal Effusions/metabolism , Clinical Trials as Topic , Gene Transfer Techniques , Guinea Pigs , Haplorhini , Intravitreal Injections/methods , Mice , Models, Animal , Nanoparticles/administration & dosage , Photoreceptor Cells, Vertebrate/drug effects , Rabbits , Rats , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Surface Properties/drug effects , Swine , Vitrectomy/methods
8.
Expert Opin Investig Drugs ; 29(3): 237-244, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31985300

ABSTRACT

Introduction: Plasma kallikrein is a  mediator of vascular leakage and inflammation. Activation of plasma kallikrein can induce features of diabetic macular edema (DME) in preclinical models. Human vitreous shows elevated plasma kallikrein levels in patients with DME. Because of the incomplete response of some patients to anti-VEGF agents, and the treatment burden associated with frequent dosing, there is still considerable need for VEGF-independent targeted pathways.Areas covered: This review covers the role of plasma kallikrein in the pathogenesis of DME and the therapeutic potential of plasma kallikrein inhibitors. It discusses early clinical studies of plasma kallikrein pathway modulation for DME, which have been associated with some improvement in visual acuity but with limited improvement in macular edema. This review also highlights KVD001, which is furthest along the development pathway, THR-149, which has recently completed a phase 1 study, and oral agents under development.Expert opinion: Plasma kallikrein inhibitors have a potential role in the treatment of DME, with mixed functional/anatomic results in early clinical trials. Given the large unmet need in DME treatment, further studies are warranted.


Subject(s)
Diabetic Retinopathy/drug therapy , Macular Edema/drug therapy , Plasma Kallikrein/antagonists & inhibitors , Animals , Diabetic Retinopathy/physiopathology , Drug Development , Drugs, Investigational/pharmacology , Humans , Macular Edema/physiopathology , Plasma Kallikrein/metabolism
9.
Transl Vis Sci Technol ; 9(13): 21, 2020 12.
Article in English | MEDLINE | ID: mdl-33364076

ABSTRACT

Purpose: This study evaluated ocular tolerability and transfectability of nonviral DNA nanoparticles (DNPs) after microneedle-based suprachoroidal (SC) administration, in comparison to subretinal (SR) administration. Methods: The DNPs consisted of a single copy of plasmid DNA with a polyubiquitin C/luciferase transcriptional cassette compacted with 10 kDa PEG-substituted lysine 30-mer peptides (CK30PEG10k). New Zealand White rabbits (n = 4 per group) received a unilateral SC injection (0.1 mL via a microneedle technique) of ellipsoid-shaped DNPs, rod-shaped DNPs, or saline (negative control). A cohort of rabbits (n = 4) also received a single unilateral SR injection (0.05 mL via a transvitreal approach) of rod-shaped DNPs. At day 7, luciferase activity was measured in the retina and retinal pigment epithelium (RPE)-choroid via bioluminescence assay. A cohort of rabbits received a SC injection of analogous DNPs to assess spread of DNP injectate in the suprachoroidal space (SCS) via optical coherent tomography and histology. Results: Suprachoroidal injection of DNPs resulted in reversible opening of the SCS circumferentially and posteriorly and was generally well tolerated, with no significant ocular examination score changes, intraocular pressure abnormalities, or changes in electroretinography amplitudes on day 7 compared to the baseline. High luciferase activity was observed in the retina and RPE-choroid of eyes that received SC DNPs (rod and ellipsoid shape) and SR DNPs (rod shape) compared to controls. The mean luciferase activity in RPE-choroid and retina was comparable between SC and SR administrations. Transfection in the RPE-choroid was approximately 10-fold higher than in the retina after either SC or SR administration of DNPs. Conclusions: Suprachoroidal and SR administration of DNPs resulted in comparable transfection of retina and RPE-choroid. Translational Relevance: Suprachoroidal delivery of DNPs offers the potential to precisely target chorioretinal tissues while avoiding surgical risks associated with SR injection, and it may offer an office-based nonsurgical gene therapy option for the treatment of retinal diseases.


Subject(s)
Nanoparticles , Retinal Pigment Epithelium , Animals , Choroid , DNA , Rabbits , Retina
10.
Expert Opin Investig Drugs ; 28(10): 861-869, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31513439

ABSTRACT

Introduction: The Tie-2/Angiopoietin pathway is a therapeutic target for the treatment of neovascular age-related macular degeneration (nAMD) and diabetic macular edema (DME). Activation of Tie-2 receptor via Ang-1 maintains vascular stability to limit exudation. Ang-2, a competitive antagonist to Ang-1, and VE-PTP, an endothelial-specific phosphatase, interfere with the Tie-2-Ang-1 axis, resulting in vascular leakage. Areas covered: Faricimab, a bispecific antibody that inhibits VEGF-A and Ang-2, is in phase 3 trials for nAMD and DME. Nesvacumab is an Ang-2 inhibitor; when coformulated with aflibercept, it failed to show benefit over aflibercept monotherapy in achieving visual gains in phase 2 studies of nAMD and DME. ARP-1536 is an intravitreally administered VE-PTP inhibitor undergoing preclinical studies. AKB-9778 is a subcutaneously administered VE-PTP inhibitor that, when combined with monthly ranibizumab, reduced DME more effectively than ranibizumab monotherapy in a phase 2 study. AKB-9778 monotherapy did not reduce diabetic retinopathy severity score compared to placebo. AXT107, currently in the preclinical phase, promotes conversion of Ang-2 into a Tie-2 agonist and blocks signaling through VEGFR2 and other receptor tyrosine-kinases. Expert opinion: Tie-2/Angiopoietin pathway modulators show promise to reduce treatment burden and improve visual outcomes in nAMD and DME, with potential to treat cases refractory to current treatment modalities.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Diabetic Retinopathy/drug therapy , Macular Degeneration/drug therapy , Angiogenesis Inhibitors/pharmacology , Angiopoietin-1/antagonists & inhibitors , Angiopoietin-2/antagonists & inhibitors , Animals , Diabetic Retinopathy/physiopathology , Humans , Macular Degeneration/physiopathology , Macular Edema/drug therapy , Macular Edema/physiopathology , Receptor, TIE-2/drug effects , Receptor, TIE-2/metabolism , Signal Transduction/drug effects
11.
Int J Pharm ; 355(1-2): 210-9, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18207340

ABSTRACT

The primary objective of this study was to investigate the expression of a specialized carrier-mediated system for folic acid and to delineate its uptake mechanism and intracellular trafficking in a human derived retinoblastoma cell line (Y-79). Uptake of [3H]Folic acid was determined at various concentrations, pH, temperatures, in the absence of sodium and chloride ions and in the presence of structural analogs, methyltetrahydro folate (MTF) and methotrexate (MTX), vitamins, membrane transport and metabolic inhibitors to delineate the mechanism of uptake. Kinetics of uptake was studied in the presence of various intracellular regulatory pathways; protein kinases A and C (PKA and PKC), protein tyrosine kinase (PTK) and calcium-calmodulin modulators. Reverse transcription polymerase chain reaction (RT-PCR) was performed to confirm the molecular identity of folate carrier systems. The uptake was found to be linear up to 30min. The rate of uptake followed saturation kinetics with apparent Km of 8.29+/-0.74nM, 17.03+/-1.98nM and 563.23+/-115.2nM and Vmax of 393.47+/-9.33, 757.58+/-26.21 and 653.17+/-31.7fmol/(minmg) protein for folic acid, MTF and MTX, respectively. The process was chloride, temperature and energy dependent but sodium and pH independent; inhibited by the structural analogs MTF and MTX but not by structurally unrelated vitamins. Membrane transport inhibitors did not affect the uptake of [3H]Folic acid, however endocytic inhibitor, colchicine, significantly inhibited the [3H]Folic acid uptake indicating the involvement of receptor mediated endocytosis process. PKC, PTK and Ca2+/calmodulin pathways appeared to play important roles in the regulation of folic acid uptake. Molecular evidence of the presence of folate receptor (FR) precursor was identified by RT-PCR analysis. This research work demonstrated, for the first time, the functional and molecular existence of a specialized high affinity carrier-mediated system for folic acid uptake, in human retinoblastoma cells.


Subject(s)
Folic Acid/administration & dosage , Folic Acid/metabolism , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Vitamins/administration & dosage , Vitamins/metabolism , Antimetabolites/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Data Interpretation, Statistical , Drug Carriers , Energy Transfer , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Transport Proteins/metabolism , Protein Kinase C/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Substrate Specificity , Temperature
12.
Int J Pharm ; 358(1-2): 168-76, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18417304

ABSTRACT

The objective of this research was to functionally characterize sodium-dependent vitamin C transporter (SVCT) in MDCK-MDR1 cells and to study the effect of substituted benzene derivatives on the intracellular accumulation of ascorbic acid (AA). Mechanism of AA uptake and transport was delineated. Uptake of [(14)C]ascorbic acid ([(14)C]AA) was studied in the absence and presence of excess unlabelled AA, anion transporter inhibitors, and a series of mono- and di-substituted benzenes. Transepithelial transport of [(14)C]AA across polarized cell membrane has been studied for the first time. Role of cellular protein kinase-mediated pathways on the regulation of AA uptake has been investigated. The cellular localizations of SVCTs were observed using confocal microscopy. Uptake of AA was found to be saturable with a K(m) of 83.2muM and V(max) of 94.2pmol/min/mg protein for SVCT1. The process was pH, sodium, temperature, and energy-dependent. It was under the regulation of cellular protein kinase C (PKC) and Ca(2+)/CaM mediated pathways. [(14)C]AA uptake was significantly inhibited in the presence of excess unlabelled AA and a series of electron-withdrawing group, i.e., halogen- and nitro-substituted benzene derivatives. AA appears to translocate across polarized cell membrane from apical to basal side (A-B) as well as basal to apical side (B-A) at a similar permeability. It appears that SVCT1 was mainly expressed on the apical side and SVCT2 may be located on both apical and basal sides. In conclusion, SVCT has been functionally characterized in MDCK-MDR1 cells. The interference of a series of electrophile-substituted benzenes on the AA uptake process may be explained by their structural similarity. SVCT may be targeted to facilitate the delivery of drugs with low bioavailability by conjugating with AA and its structural analogs. MDCK-MDR1 cell line may be utilized as an in vitro model to study the permeability of AA conjugated prodrugs.


Subject(s)
Ascorbic Acid/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Biological Transport, Active , Cell Line , Cell Membrane Permeability , Data Interpretation, Statistical , Dogs , Energy Metabolism/physiology , Humans , Hydrogen-Ion Concentration , Microscopy, Confocal , Signal Transduction/physiology , Sodium/physiology , Sodium-Coupled Vitamin C Transporters , Substrate Specificity
13.
Int J Pharm ; 336(2): 233-40, 2007 May 24.
Article in English | MEDLINE | ID: mdl-17207946

ABSTRACT

Saquinavir (SQV) was the first human immuno-virus-1 (HIV-1) protease inhibitor approved by FDA. However, P-glycoprotein (P-gp), an efflux pump limits its oral and brain bioavailabilities. The objective of this study is to investigate whether prodrug modification of SQV to dipeptide prodrugs Valine-Valine-Saquinavir (Val-Val-SQV) and Glycine-Valine-Saquinavir (Gly-Val-SQV) targeting intestinal peptide transporter can enhance intestinal permeability of SQV by circumventing P-gp mediated efflux. Single pass intestinal perfusion experiments in rat jejunum were performed to calculate the absorption rate constant and intestinal permeability of SQV, Val-Val-SQV and Gly-Val-SQV. Equimolar concentration (25 microM) of SQV, Val-Val-SQV and Gly-Val-SQV were employed in the perfusion studies. Perfusion experiments were also carried out in the presence of cyclosporine (10 microM) and glycyl-sarcosine (20 mM). Absorption rate constants in rat jejunum (ka) for SQV, Val-Val-SQV and Gly-Val-SQV were found to be 14.1+/-3.4x10(-3), 65.8+/-4.3x10(-3), and 25.6+/-5.7x10(-3) min(-1), respectively. Enhanced absorption of Val-Val-SQV and Gly-Val-SQV relative to SQV can be attributed to their translocation by the peptide transporter in the jejunum. Significant permeability enhancement of SQV across rat jejunum was observed in the presence of cyclosporine 10 microM (P-gp inhibitor). However, permeability of Val-Val-SQV was unchanged in the presence of cyclosporine suggesting lack of any interaction of the prodrug with efflux pump. Intestinal absorption of Val-Val-SQV was significantly inhibited in the presence of gly-sar indicating the involvement of peptide transporter in intestinal absorption. In conclusion, peptide transporter targeted prodrug modification of P-gp substrates could lead to shielding of these drug molecules from efflux pumps.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , Prodrugs/pharmacokinetics , Saquinavir/analogs & derivatives , Saquinavir/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Biological Availability , Biological Transport , Drug Stability , HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/chemistry , Intestinal Absorption , Jejunum/metabolism , Male , Permeability , Prodrugs/chemical synthesis , Prodrugs/chemistry , Rats , Rats, Sprague-Dawley , Saquinavir/chemical synthesis , Saquinavir/chemistry
14.
J Ocul Pharmacol Ther ; 23(4): 321-34, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17803430

ABSTRACT

PURPOSE: The overall aim of this research work was to evaluate a series of dipeptide monoester prodrugs of an antiviral agent, ganciclovir (GCV), for oligopeptide transporter-targeted transscleral drug delivery to rabbit retina. METHODS: The permeability and enzymatic hydrolysis of dipeptide monoester GCV prodrugs were evaluated using freshly excized rabbit retinal pigment epithelium (RPE)-choroidsclera (RCS) and sclera tissue preparations. Affinity and transport mechanism of these prodrugs and their translocation across RCS were investigated through competitive inhibition studies of [(3)H]glycylsarcosine with the prodrugs. RESULTS: The transport of glycylsarcosine was found to be saturable (K(m) = 1.21 +/- 0.41 mM, V(max) = 15.89 +/- 1.54 pmoles/min/cm(2)), pH, temperature, and energy dependant. Dipeptides, angiotensin converting enzyme inhibitors, and a beta-lactum antibiotic strongly inhibited the transport of glycylsarcosine indicating the functional presence of oligopeptide transport (OPT) system on the RPE. Dipeptide prodrugs (valine-valine-GCV, glycine-valine-GCV, and tyrosine-valine-GCV), and valine-GCV demonstrate a high enzymatic stability and affinity toward the retinal OPT system. The transport of the prodrugs was significantly inhibited ( approximately 50%) in the presence of glycylsarcosine. The rank order of scleral permeability was Gly-Val-GCV approximately GCV>Val-GCV>Tyr-Val-GCV approximately Val-Val-GCV. The RCS permeability values of Val-GCV (3.29 +/- 0.09 x 10(6)cm/s), Val-Val-GCV (4.14 +/- 0.33 x 10(6)cm/s), Gly-Val-GCV (3.40 +/- 0.47 x 10(6)cm/s) and Tyr-Val-GCV (3.08 +/- 0.29 x 10(6)cm/s) were two-fold higher than that of GCV (1.61 +/- 0.06 x 10(6)cm/s). CONCLUSIONS: The dipeptide monoester GCV prodrugs, owning to higher lipophilicity and OPT-mediated translocation across RPE, appear to be promising candidates in the treatment of ocular cytomegalovirus infections following an episcleral administration.


Subject(s)
Antiviral Agents/pharmacokinetics , Drug Delivery Systems , Ganciclovir/pharmacokinetics , Retina/metabolism , Animals , Antiviral Agents/administration & dosage , Biological Transport , Carrier Proteins/metabolism , Choroid/metabolism , Dipeptides/administration & dosage , Dipeptides/pharmacokinetics , Ganciclovir/administration & dosage , Ganciclovir/analogs & derivatives , In Vitro Techniques , Male , Oligopeptides/metabolism , Pigment Epithelium of Eye/metabolism , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Rabbits , Sclera/metabolism , Structure-Activity Relationship
15.
SLAS Discov ; 22(10): 1239-1245, 2017 12.
Article in English | MEDLINE | ID: mdl-28787579

ABSTRACT

Matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) is becoming an important technology to determine the distribution of drugs and their metabolites in the tissue of preclinical species after dosing. Interest in IMS is growing in the ophthalmology field, but little work to this point has been done to investigate ocular drug transit using this technology. Information on where and how a drug is distributing through the eye is important in understanding efficacy and whether it is reaching the desired target tissue. For this study, ocular distribution of brimonidine was investigated in rabbits following topical administration. Brimonidine has been shown to lower intraocular pressure and is approved to treat glaucoma, the second leading cause of blindness in the world. We have developed IMS methods to assess transit of topically administered brimonidine from the anterior to the posterior segment of rabbit eyes. Using IMS, brimonidine was detected in the cornea, aqueous humor, iris, and posterior segments of the eye. The distribution of brimonidine suggests that the route of transit following topical administration is mainly through the uvea-scleral route. This study demonstrates that IMS can be applied to assess ocular transit and distribution of topically administered drugs.


Subject(s)
Brimonidine Tartrate/pharmacology , Eye/drug effects , Imaging, Three-Dimensional , Mass Spectrometry , Animals , Rabbits , Signal Processing, Computer-Assisted , Time Factors
16.
J Med Chem ; 60(13): 5717-5735, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28621538

ABSTRACT

The highly specific S1 serine protease factor D (FD) plays a central role in the amplification of the complement alternative pathway (AP) of the innate immune system. Genetic associations in humans have implicated AP activation in age-related macular degeneration (AMD), and AP dysfunction predisposes individuals to disorders such as paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). The combination of structure-based hit identification and subsequent optimization of the center (S)-proline-based lead 7 has led to the discovery of noncovalent reversible and selective human factor D (FD) inhibitors with drug-like properties. The orally bioavailable compound 2 exerted excellent potency in 50% human whole blood in vitro and blocked AP activity ex vivo after oral administration to monkeys as demonstrated by inhibition of membrane attack complex (MAC) formation. Inhibitor 2 demonstrated sustained oral and ocular efficacy in a model of lipopolysaccharide (LPS)-induced systemic AP activation in mice expressing human FD.


Subject(s)
Complement Factor D/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Proline/analogs & derivatives , Proline/pharmacology , Administration, Oral , Animals , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/immunology , Complement Factor D/immunology , Complement Membrane Attack Complex/antagonists & inhibitors , Complement Membrane Attack Complex/immunology , Female , Haplorhini , Humans , Macaca fascicularis , Macular Degeneration/drug therapy , Macular Degeneration/immunology , Male , Mice , Proline/administration & dosage , Proline/pharmacokinetics
17.
Nat Commun ; 8: 14837, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332616

ABSTRACT

Protein drugs that neutralize vascular endothelial growth factor (VEGF), such as aflibercept or ranibizumab, rescue vision in patients with retinal vascular diseases. Nonetheless, optimal visual outcomes require intraocular injections as frequently as every month. Here we report a method to extend the intravitreal half-life of protein drugs as an alternative to either encapsulation or chemical modifications with polymers. We combine a 97-amino-acid peptide of human origin that binds hyaluronan, a major macromolecular component of the eye's vitreous, with therapeutic antibodies and proteins. When administered to rabbit and monkey eyes, the half-life of the modified proteins is increased ∼3-4-fold relative to unmodified proteins. We further show that prototype long-acting anti-VEGF drugs (LAVAs) that include this peptide attenuate VEGF-induced retinal changes in animal models of neovascular retinal disease ∼3-4-fold longer than unmodified drugs. This approach has the potential to reduce the dosing frequency associated with retinal disease treatments.


Subject(s)
Bevacizumab/administration & dosage , Ranibizumab/administration & dosage , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Retinal Diseases/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Animals , Bevacizumab/chemistry , Bevacizumab/pharmacokinetics , Disease Models, Animal , Female , Half-Life , Humans , Hyaluronic Acid/chemistry , Intravitreal Injections , Macaca fascicularis , Male , Rabbits , Ranibizumab/chemistry , Ranibizumab/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Retinal Diseases/metabolism
18.
Int J Pharm ; 312(1-2): 43-52, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16459033

ABSTRACT

The objective of this research was to investigate the presence of a specialized carrier-mediated system for biotin and delineate uptake mechanism and intracellular trafficking of biotin in the human derived retinoblastoma cell line (Y-79). Human derived retinoblastoma cell line, Y-79, was used for uptake studies. Uptake of [3H]Biotin was determined at various concentrations, pH, temperatures, in the absence of sodium and in the presence of other vitamins and metabolic inhibitors to delineate the mechanism of uptake. Uptake was determined in the presence of various intracellular regulatory pathways (protein kinase A & C, protein tyrosine kinase and calcium-calmodulin) modulators. Reverse transcription polymerase chain reaction (RT-PCR) was performed to confirm the molecular identity of human sodium-dependent multivitamin transporter (hSMVT). Uptake of [3H]Biotin in Y-79 cells were found to be saturable at micromolar concentration range, with apparent Km of 8.53 microM and Vmax of 14.12 pmol/min/mg protein, but linear at nanomolar concentration range. Uptake was sodium, pH, temperature and energy-dependent, but chloride independent; inhibited by the structural analogue desthiobiotin, pantothenic acid and lipoic acid at milimolar concentrations and not at nanomolar concentrations. Uptake of [3H]Biotin was trans-stimulated by the intracellular biotin. Ca2+/calmodulin pathways appeared to play important roles in the regulation of riboflavin uptake in Y-79 cells via significant reduction in Vmax (66%) and Km (28%) of the uptake process. A human sodium-dependant multivitamin transporter, hSMVT, was identified by RT-PCR in Y-79. These studies demonstrated for the first time the existence of a human sodium dependant multivitamin transporter (hSMVT), a specialized carrier-mediated system for biotin uptake, in human derived retinoblastoma cells.


Subject(s)
Biotin/metabolism , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Symporters/physiology , Biological Transport , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Reverse Transcriptase Polymerase Chain Reaction , Temperature
19.
Curr Eye Res ; 31(5): 415-26, 2006 May.
Article in English | MEDLINE | ID: mdl-16714233

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the implication of an ex vivo model for carrier-mediated retinal drug delivery using an Ussing chamber system. METHODS: Dutch Belted Pigmented rabbits weighing 2-2.5 kg were used in these studies. Excised posterior segment tissues (RPE-choroid-sclera and sclera), mounted on the Ussing chamber, were used as an ex vivo model. Transport studies were carried out across sclera and RPE-choroid-sclera (RCS) tissue preparations in the sclera to retina (S --> R) and retina to sclera (R --> S) directions for 3 hr at 37 degrees C. The model was validated by permeability studies with paracellular and transcellular markers ([(3)H]mannitol and [(14)C]diazepam, respectively), tissue viability studies (bioelectrical and biochemical assays), and tissue histology and electron microscopy studies. Functional presence of a carrier-mediated transport system for folic acid (folate receptor alpha) was investigated on the basolateral side of the rabbit retina. RESULTS: Results from bioelectrical, biochemical, and histological evaluation of tissue provide evidence that the RCS tissue preparation remains viable during the period of transport study. Permeability values of [(3)H]mannitol across sclera were 4.18 +/- 1.09 x 10(- 5) cm/s (R --> S) and 4.11 +/- 1.09 x 10(- 5) cm/s (S --> R) and across RCS were 1.77 +/- 0.31 x 10(- 5) cm/s (S --> R) and 1.60 +/- 0.19 x 10(- 5) cm/s (R --> S). Permeability values of [(14)C]diazepam across sclera were 2.37 +/- 0.38 x 10(- 5) cm/s (R --> S) and 2.70 +/- 0.70 x 10(- 5) cm/s (S --> R) and across RCS were 3.12 +/- 0.12 x 10(- 5) cm/s (R --> S) and 2.77 +/- 0.25 x 10(- 5)cm/s (S --> R). The rate of [(3)H]folic acid transport across RCS was found to be significantly higher in the S -->R direction (16.34 +/- 0.94 fmoles min(-1) cm(-2)) as compared with R --> S direction (9.38 +/- 1.44 fmoles min(-1) cm(-2)) and nearly 10-fold higher across sclera as compared with RCS in both directions. Transport of [(3)H]folic acid was found to be pH and temperature dependent and was inhibited by 44.5%, 35.1%, and 50.3% in the presence of unlabeled folic acid, 5-methyltetrahydrofolate (MTF), and Methotrexate (MTX). CONCLUSIONS: The RCS tissue preparation mounted on the Ussing chamber system, an ex vivo model, can be a useful tool for identification and characterization of carrier-mediated systems present on RPE (a major barrier for retinal drug delivery) and to study carrier-mediated retinal drug delivery via prodrug derivatization.


Subject(s)
Carrier Proteins/metabolism , Drug Delivery Systems , Folic Acid/metabolism , Pigment Epithelium of Eye/metabolism , Receptors, Cell Surface/metabolism , Retina/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Cell Survival , Choroid/metabolism , Diazepam/metabolism , Diffusion Chambers, Culture , Electric Conductivity , Folate Receptors, GPI-Anchored , Hydrogen-Ion Concentration , Mannitol/metabolism , Membrane Potentials/physiology , Methotrexate/pharmacology , Models, Biological , Rabbits , Sclera/metabolism , Temperature , Tetrahydrofolates/physiology
20.
J Ocul Pharmacol Ther ; 22(4): 231-41, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16910863

ABSTRACT

PURPOSE: The aim of this study was to determine vitreal pharmacokinetics of a series of dipeptide monoester ganciclovir (GCV) prodrugs and to study their interaction with the retinal peptide transporter. METHODS: New Zealand albino male rabbits were selected as the animal model. Ocular microdialysis technique was employed to delineate the pharmacokinetics of GCV, L-valine-GCV and dipeptide monoester GCV prodrugs (L-valine-L-valine, L-tyrosine-L-valine, and L-glycine- L-valine) following intravitreal administration. RESULTS: Val-GCV and Val-Val-GCV inhibited retinal uptake of [3H]Gly-Sar by 43% and 37%, respectively, suggesting that these prodrugs may be substrates of the retinal peptide transport system. Val-GCV and Gly-Val-GCV were observed to be the most stable GCV prodrugs in vitreous humor. All GCV prodrugs were rapidly converted to GCV in retinal homogenates. Vitreal pharmacokinetic studies suggest that Val-GCV and Val-Val-GCV are rapidly eliminated from the vitreous chamber, compared to GCV, whereas Gly-Val-GCV is eliminated at a much slower rate. Retinal GCV concentrations generated from all three prodrugs, at the end of 5 h, were almost equivalent and were almost twice that following intravitreal administration of GCV. Gly-Pro, however, did not demonstrate any effect on retinal uptake of Val-GCV or Gly-Val-GCV. CONCLUSIONS: Considering retinal GCV concentrations generated and vitreal pharmacokinetic profiles, Gly-Val-GCV appears to be a lead candidate for further in vivo evaluation against human cytomegalovirus (HCMV) retinitis.


Subject(s)
Antiviral Agents/pharmacokinetics , Dipeptides/pharmacokinetics , Ganciclovir/pharmacokinetics , Prodrugs/pharmacokinetics , Vitreous Body/metabolism , Animals , Biotransformation , Choroid/metabolism , Chromatography, High Pressure Liquid , Ganciclovir/analogs & derivatives , Male , Rabbits , Retina/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL