1
|
Heljak MK, Cesur S, Ilhan E, Swieszkowski W, Gunduz O, Kijeńska-Gawrońska E. In silico evaluation of corneal patch eluting anti-VEGF agents concept. Eur J Pharm Biopharm 2024; 204:114494. [PMID: 39255920 DOI: 10.1016/j.ejpb.2024.114494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
This study introduces a novel approach utilizing a temporary drug-eluting hydrogel corneal patch to prevent neovascularization, alongside a numerical predictive tool for assessing the release and transport kinetics of bevacizumab (BVZ) after the keratoplasty. A key focus was investigating the impact of tear film clearance on the release kinetics and drug transport from the designed corneal patch. The proposed tear drug clearance model incorporates the physiological mechanism of lacrimal flow (tear turnover), distinguishing itself from previous models. Validation against experimental data confirms the model's robustness, despite limitations such as a 2D axisymmetrical framework and omission of blink frequency and saccadic eye movements potential effects. Analysis highlights the significant influence of lacrimal flow on ocular drug transport, with the corneal patch extending BVZ residence time compared to topical administration. This research sets the stage for exploring multi-layer drug-eluting corneal patches as a promising therapeutic strategy in ocular health.
Collapse
Affiliation(s)
- Marcin K Heljak
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Poland.
| | - Sumeyye Cesur
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Turkiye; Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Turkiye
| | - Elif Ilhan
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Turkiye
| | - Wojciech Swieszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Poland
| | - Oguzhan Gunduz
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Turkiye; Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Turkiye
| | - Ewa Kijeńska-Gawrońska
- Centre for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poland.
| |
Collapse
|
2
|
Lamirande P, Gaffney EA, Gertz M, Maini PK, Crawshaw JR, Caruso A. A First-Passage Model of Intravitreal Drug Delivery and Residence Time-Influence of Ocular Geometry, Individual Variability, and Injection Location. Invest Ophthalmol Vis Sci 2024; 65:21. [PMID: 39412819 PMCID: PMC11488524 DOI: 10.1167/iovs.65.12.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/12/2024] [Indexed: 10/20/2024] Open
Abstract
Purpose Standard of care for various retinal diseases involves recurrent intravitreal injections. This motivates mathematical modeling efforts to identify influential factors for ocular drug residence time, aiming to minimize administration frequency. We sought to describe the vitreal diffusion of therapeutics in nonclinical species frequently used during drug development assessments. In human eyes, we investigated the impact of variability in vitreous cavity size and eccentricity, and in injection location, on drug disposition. Methods Using a first-passage time approach, we modeled the transport-controlled distribution of two standard therapeutic protein formats (Fab and IgG) and elimination through anterior and posterior pathways. Anatomical three-dimensional geometries of mouse, rat, rabbit, cynomolgus monkey, and human eyes were constructed using ocular images and biometry datasets. A scaling relationship was derived for comparison with experimental ocular half-lives. Results Model simulations revealed a dependence of residence time on ocular size and injection location. Delivery to the posterior vitreous resulted in increased vitreal half-life and retinal permeation. Interindividual variability in human eyes had a significant influence on residence time (half-life range of 5-7 days), showing a strong correlation to axial length and vitreal volume. Anterior exit was the predominant route of drug elimination. Contribution of the posterior pathway displayed a 3% difference between protein formats but varied between species (10%-30%). Conclusions The modeling results suggest that experimental variability in ocular half-life is partially attributed to anatomical differences and injection site location. Simulations further suggest a potential role of the posterior pathway permeability in determining species differences in ocular pharmacokinetics.
Collapse
Affiliation(s)
- Patricia Lamirande
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
| | - Eamonn A. Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
| | - Michael Gertz
- Pharmaceutical Sciences, Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
| | - Jessica R. Crawshaw
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Oxford, United Kingdom
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Antonello Caruso
- Pharmaceutical Sciences, Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| |
Collapse
|
3
|
Sadeghi A, Subrizi A, Del Amo EM, Urtti A. Mathematical Models of Ocular Drug Delivery. Invest Ophthalmol Vis Sci 2024; 65:28. [PMID: 39287588 PMCID: PMC11412384 DOI: 10.1167/iovs.65.11.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Drug delivery is an important factor for the success of ocular drug treatment. However, several physical, biochemical, and flow-related barriers limit drug exposure of anterior and posterior ocular target tissues during drug treatment via topical, subconjunctival, intravitreal, or systemic routes. Mathematical models encompass various barriers so that their joint influence on pharmacokinetics (PKs) can be simulated in an integrated fashion. The models are useful in predicting PKs and even pharmacodynamics (PDs) of administered drugs thereby fostering development of new drug molecules and drug delivery systems. Furthermore, the models are potentially useful in interspecies translation and probing of disease effects on PKs. In this review article, we introduce current modeling methods (noncompartmental analyses, compartmental and physiologically based PK models, and finite element models) in ocular PKs and related drug delivery. The roles of top-down models and bottom-up simulations are discussed. Furthermore, we present some future challenges, such as modeling of intra-tissue distribution, prediction of drug responses, quantitative systems pharmacology, and possibilities of artificial intelligence.
Collapse
Affiliation(s)
- Amir Sadeghi
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Eva M Del Amo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
4
|
Chowdhury JM, Chacin Ruiz EA, Ohr MP, Swindle-Reilly KE, Ford Versypt AN. Computer Modeling of Bevacizumab Drug Distribution after Intravitreal Injection in Rabbit and Human Eyes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.05.539491. [PMID: 37215026 PMCID: PMC10197542 DOI: 10.1101/2023.05.05.539491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Age-related macular degeneration (AMD) is a progressive eye disease that causes loss of central vision and has no cure. Wet AMD is the late neovascular form treated with vascular endothelial growth factor (VEGF) inhibitors. VEGF is the critical driver of wet AMD. One common off-label anti-VEGF drug used in AMD treatment is bevacizumab. Experimental efforts have been made to investigate the pharmacokinetic (PK) behavior of bevacizumab in vitreous and aqueous humor. Still, the quantitative effect of elimination routes and drug concentration in the macula are not well understood. In this work, we developed two spatial models representing rabbit and human vitreous to better understand the PK behavior of bevacizumab. This study explores different cases of drug elimination and the effects of injection location on drug concentration profiles. The models are validated by comparing them with experimental data. Our results suggest that anterior elimination is dominant for bevacizumab clearance from rabbit vitreous, whereas both anterior and posterior elimination have similar importance in drug clearance from the human vitreous. Furthermore, results indicate that drug injections closer to the posterior segment of the vitreous help maintain relevant drug concentrations for longer, improving bevacizumab duration of action at the fovea, which is located at the center of the macula. The rabbit and human models predict bevacizumab concentration in the vitreous and fovea, enhancing knowledge and understanding of wet AMD treatment.
Collapse
|
5
|
Hammid A, Honkakoski P. Ocular Drug-Metabolizing Enzymes: Focus on Esterases. Drug Metab Rev 2024:1-23. [PMID: 38888291 DOI: 10.1080/03602532.2024.2368247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Affiliation(s)
- Anam Hammid
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne3, FI-70210 Kuopio, Finland
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne3, FI-70210 Kuopio, Finland
| |
Collapse
|
6
|
Merivaara A, Puranen J, Sadeghi A, Zashikhina N, Pirskanen L, Lajunen T, Terasaki T, Auriola S, Vellonen KS, Urtti A. Barcode lipids for absolute quantitation of liposomes in ocular tissues. J Control Release 2024; 370:1-13. [PMID: 38615893 DOI: 10.1016/j.jconrel.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/14/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Lipid-based drug formulations are promising systems for improving delivery of drugs to ocular tissues, such as retina. To develop lipid-based systems further, an improved understanding of their pharmacokinetics is required, but high-quality in vivo experiments require a large number of animals, raising ethical and economic questions. In order to expedite in vivo kinetic testing of lipid-based systems, we propose a barcode approach that is based on barcoding liposomes with non-endogenous lipids. We developed and evaluated a liquid-chromatography-mass spectrometry method to quantify many liposomes simultaneously in aqueous humor, vitreous, and neural retina at higher than ±20% precision and accuracy. Furthermore, we showed in vivo suitability of the method in pharmacokinetic evaluation of six different liposomes after their simultaneous injection into the rat vitreal cavity. We calculated pharmacokinetic parameters in vitreous and aqueous humor, quantified liposome concentrations in the retina, and quantitated retinal distribution of the liposomes in the rats. Compared to individual injections of the liposome formulations, the barcode-based study design enabled reduction of animal numbers from 72 to 12. We believe that the proposed approach is reliable and will reduce and refine ocular pharmacokinetic experiments with liposomes and other lipid-based systems.
Collapse
Affiliation(s)
- Arto Merivaara
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland.
| | - Jooseppi Puranen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland
| | - Amir Sadeghi
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland
| | - Natalia Zashikhina
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland
| | - Lea Pirskanen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland
| | - Tatu Lajunen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki 00014, Finland
| | - Tetsuya Terasaki
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland.
| | - Kati-Sisko Vellonen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, Kuopio 70210, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
7
|
Lee S, Kim SN, Lee C, Choy YB, Im CH. Multi-physics simulations for investigating the effect of electrode conditions on transscleral ocular iontophoresis for particulate drug delivery into ocular tissues. Biomed Eng Lett 2024; 14:439-450. [PMID: 38645594 PMCID: PMC11026336 DOI: 10.1007/s13534-024-00359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 04/23/2024] Open
Abstract
Purpose Transscleral ocular iontophoresis has been proposed to deliver charged particulate drugs to ocular tissues effectively by transmitting a weak electrical current through the sclera. The electric fields formed are influenced by the electrode conditions, thus affecting the amount of particulate drugs delivered to the ocular tissues via iontophoresis. Computational simulation is widely used to simulate drug concentrations in the eye; therefore, reflecting the characteristics of the drugs in living tissues to the simulations is important for a more precise estimation of drug concentration. In this study, we investigated the effect of electrode conditions (location and size) on the efficacy of transscleral iontophoresis. Methods We first determined the simulation parameters based on the comparison of the amount of drug in the sclera in the simulation and in vivo experimental results. The injection of the negatively charged nanoparticles into the cul-de-sac of the lower eyelid was simulated. The active electrode (cathode) was attached to the skin immediately above the injection site, while the return electrode (anode) was placed over the eyebrow. The drug concentration distribution in the eye, based on either the location or size of each electrode, was evaluated using the finite element method with the estimated simulation parameters. Results Our results indicate that drug permeability varies depending on the location and the size of the electrodes. Conclusion Our findings demonstrate that the determination of optimal electrode conditions is necessary to enhance the effectiveness of transscleral iontophoresis. Supplementary Information The online version contains supplementary material available at 10.1007/s13534-024-00359-2.
Collapse
Affiliation(s)
- Sangjun Lee
- Department of Biomedical Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong- gu, Seoul, 133-791 Republic of Korea
- Department of Electronic Engineering, Hanyang University, Seoul, Republic of Korea
| | - Se-Na Kim
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Chany Lee
- Department of Structure & Function of Neural Network, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Young Bin Choy
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang-Hwan Im
- Department of Biomedical Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong- gu, Seoul, 133-791 Republic of Korea
- Department of Electronic Engineering, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
8
|
Ruffini A, Casalucci A, Cara C, Ethier CR, Repetto R. Drug Distribution After Intravitreal Injection: A Mathematical Model. Invest Ophthalmol Vis Sci 2024; 65:9. [PMID: 38568619 PMCID: PMC10996986 DOI: 10.1167/iovs.65.4.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Purpose Intravitreal injection of drugs is commonly used for treatment of chorioretinal ocular pathologies, such as age-related macular degeneration. Injection causes a transient increase in the intraocular volume and, consequently, of the intraocular pressure (IOP). The aim of this work is to investigate how intravitreal flow patterns generated during the post-injection eye deflation influence the transport and distribution of the injected drug. Methods We present mathematical and computational models of fluid motion and mass transport in the vitreous chamber during the transient phase after injection, including the previously unexplored effects of globe deflation as ocular volume decreases. Results During eye globe deflation, significant fluid velocities are generated within the vitreous chamber, which can possibly contribute to drug transport. Pressure variations within the eye globe are small compared to IOP. Conclusions Even if significant fluid velocities are generated in the vitreous chamber after drug injection, these are found to have negligible overall effect on drug distribution.
Collapse
Affiliation(s)
- Alessia Ruffini
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, Genoa, Italy
| | - Alessia Casalucci
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, Genoa, Italy
| | - Caterina Cara
- Department of Civil, Environmental and Architectural Engineering, University of Padua, Padua, Italy
| | - C Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, United States
| | - Rodolfo Repetto
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, Genoa, Italy
| |
Collapse
|
9
|
Heljak MK, Swieszkowski W. Investigating bevacizumab and its fragments sustained release from intravitreal administrated PLGA Microspheres: A modeling approach. Eur J Pharm Biopharm 2023; 193:285-293. [PMID: 37984593 DOI: 10.1016/j.ejpb.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Intravitreal administrated bevacizumab has emerged as an effective antibody for suppressing VEGF expression in age-related macular degeneration (AMD) therapy. This study discusses certain issues related to the sustained release of bevacizumab from intravitreal poly(lactic-co-glycolic acid) (PLGA) microspheres. A computational model elucidating the ocular kinetics of bevacizumab is demonstrated, wherein the release of the drug from PLGA microspheres is modeled using the Koizumi approach, complemented by an empirical model that links the kinetics of bevacizumab release to a size-dependent hydrolytic degradation of the drug-loaded polymeric microparticles. The results of the simulation were then rigorously validated against experimental data. The as-developed model proved remarkably accurate in predicting the time-concentration profiles obtained following the intravitreal injection of PLGA microspheres of significantly different sizes. Notably, the time-concentration profiles of bevacizumab in distinct ocular tissues were almost unaffected by the size of the intravitreally administered PLGA microparticles. Furthermore, the model successfully predicted the retinal concentration of bevacizumab and its fragments (e.g., ranibizumab) administrated in the form of a solution. As such, this model for drug sustained release and ocular transport holds tremendous potential for facilitating the reliable evaluation of planned anti-VEGF therapies.
Collapse
Affiliation(s)
- Marcin K Heljak
- Faculty of Materials Science and Engineering, Warsaw University of Technology, ul. Wołoska 141, 02-507 Warsaw, Poland
| | - Wojciech Swieszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, ul. Wołoska 141, 02-507 Warsaw, Poland
| |
Collapse
|
10
|
Chacin Ruiz EA, Swindle-Reilly KE, Ford Versypt AN. Experimental and mathematical approaches for drug delivery for the treatment of wet age-related macular degeneration. J Control Release 2023; 363:464-483. [PMID: 37774953 PMCID: PMC10842193 DOI: 10.1016/j.jconrel.2023.09.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/01/2023]
Abstract
Several chronic eye diseases affect the posterior segment of the eye. Among them age-related macular degeneration can cause vision loss if left untreated and is one of the leading causes of visual impairment in the world. Most treatments are based on intravitreally injected therapeutics that inhibit the action of vascular endothelial growth factor. However, due to the need for monthly injections, this method is associated with poor patient compliance. To address this problem, numerous drug delivery systems (DDSs) have been developed. This review covers a selection of particulate systems, non-stimuli responsive hydrogels, implants, and composite systems that have been developed in the last few decades. Depending on the type of DDS, polymer material, and preparation method, different mechanical properties and drug release profiles can be achieved. Furthermore, DDS development can be optimized by implementing mathematical modeling of both drug release and pharmacokinetic aspects. Several existing mathematical models for diffusion-controlled, swelling-controlled, and erosion-controlled drug delivery from polymeric systems are summarized. Compartmental and physiologically based models for ocular drug transport and pharmacokinetics that have studied drug concentration profiles after intravitreal delivery or release from a DDS are also reviewed. The coupling of drug release models with ocular pharmacokinetic models can lead to obtaining much more efficient DDSs for the treatment of age-related macular degeneration and other diseases of the posterior segment of the eye.
Collapse
Affiliation(s)
- Eduardo A Chacin Ruiz
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Katelyn E Swindle-Reilly
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA; Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, USA
| | - Ashlee N Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA; Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY, USA.
| |
Collapse
|
11
|
Puranen J, Ranta VP, Ruponen M, Urtti A, Sadeghi A. Quantitative intravitreal pharmacokinetics in mouse as a step towards inter-species translation. Exp Eye Res 2023; 235:109638. [PMID: 37657528 DOI: 10.1016/j.exer.2023.109638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/17/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Although mouse models are widely used in retinal drug development, pharmacokinetics in mouse eye is poorly understood. In this study, we applied non-invasive in vivo fluorophotometry to study pharmacokinetics of intravitreal fluorescein sodium (molecular weight 0.38 kDa) and fluorescein isothiocyanate-dextran (FD-150; molecular weight 150 kDa) in mice. Intravitreal half-lives of fluorescein and FD-150 in mouse eyes were 0.53 ± 0.06 h and 2.61 ± 0.86 h, respectively. These values are 8-230 times shorter than the elimination half-lives of similar compounds in the human vitreous. The apparent volumes of distribution in the mouse vitreous were close to the anatomical volume of the mouse vitreous (FD-150, 5.1 μl; fluorescein, 9.6 μl). Dose scaling factors were calculated from mouse to rat, rabbit, monkey and human translation. Based on pharmacokinetic modelling and compound concentrations in the vitreous and anterior chamber, fluorescein is mainly eliminated posteriorly across blood-retina barrier, but FD-150 is cleared via aqueous humour outflow. The results of this study improve the knowledge of intravitreal pharmacokinetics in mouse and facilitate inter-species scaling in ocular drug development.
Collapse
Affiliation(s)
- Jooseppi Puranen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland.
| | - Veli-Pekka Ranta
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Marika Ruponen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00790, Finland
| | - Amir Sadeghi
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland.
| |
Collapse
|
12
|
Naware S, Bussing D, Shah DK. Translational physiologically-based pharmacokinetic model for ocular disposition of monoclonal antibodies. J Pharmacokinet Pharmacodyn 2023:10.1007/s10928-023-09881-9. [PMID: 37558929 DOI: 10.1007/s10928-023-09881-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
We have previously published a PBPK model comprising the ocular compartment to characterize the disposition of monoclonal antibodies (mAbs) in rabbits. While rabbits are commonly used preclinical species in ocular research, non-human primates (NHPs) have the most phylogenetic resemblance to humans including the presence of macula in the eyes as well as higher sequence homology. However, their use in ocular research is limited due to the strict ethical guidelines. Similarly, in humans the ocular samples cannot be collected except for the tapping of aqueous humor (AH). Therefore, we have translated this rabbit model to monkeys and human species using literature-reported datasets. Parameters describing the tissue volumes, physiological flows, and FcRn-binding were obtained from the literature, or estimated by fitting the model to the data. In the monkey model, the values for the rate of lysosomal degradation for antibodies (Kdeg), intraocular reflection coefficients (σaq, σret, σcho), bidirectional rate of fluid circulation between the vitreous chamber and the aqueous chamber (QVA), and permeability-surface area product of lens (PSlens) were estimated; and were found to be 31.5 h-1, 0.7629, 0.6982, 0.9999, 1.64 × 10-5 L/h, and 4.62 × 10-7 L/h, respectively. The monkey model could capture the data in plasma, aqueous humor, vitreous humor and retina reasonably well with the predictions being within twofold of the observed values. For the human model, only the value of Kdeg was estimated to fit the model to the plasma pharmacokinetics (PK) of mAbs and was found to be 24.4 h-1 (4.14%). The human model could also capture the ocular PK data reasonably well with the predictions being within two- to threefold of observed values for the plasma, aqueous and vitreous humor. Thus, the proposed framework can be used to characterize and predict the PK of mAbs in the eye of monkey and human species following systemic and intravitreal administration. The model can also facilitate the development of new antibody-based therapeutics for the treatment of ocular diseases as well as predict ocular toxicities of such molecules following systemic administration.
Collapse
Affiliation(s)
- Sanika Naware
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York, University at Buffalo 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA
| | - David Bussing
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York, University at Buffalo 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York, University at Buffalo 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA.
| |
Collapse
|
13
|
Toffoletto N, Saramago B, Serro AP, Chauhan A. A Physiology-Based Mathematical Model to Understand Drug Delivery from Contact Lenses to the Back of the Eye. Pharm Res 2023; 40:1939-1951. [PMID: 37498499 PMCID: PMC10447275 DOI: 10.1007/s11095-023-03560-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/02/2023] [Indexed: 07/28/2023]
Abstract
OBJECTIVE Therapeutic contact lenses, able to store drug and deliver it to the eye surface in a sustained fashion, gained interest as an effective and patient-friendly alternative to eye drops. Recent animal studies also demonstrated the presence of therapeutic drug levels in the back of the eye after wearing drug-loaded contact lenses, thus opening the possibility of treating the posterior segment without need of invasive intraocular injections. The drug pathways from contact lenses to the back of the eye require further investigation. METHODS A mechanistic mathematical model was developed to evaluate the drug concentration over time in the tears, sclera and choroid, retina, aqueous humor and vitreous humor after the application of a therapeutic contact lens. The main drug transport mechanisms of the eye and the barrier properties of the different tissues were included in the model. Validation was performed by comparison with experimental data in literature. RESULTS The model predictions of drug concentration over time reflected the experimental data both in the anterior and posterior segment of the eye. The model can differentiate between contributions to transport from different pathways. CONCLUSIONS The model constitutes a first step towards the possibility of predicting the ocular drug distribution and the treatment efficacy in the early stage of contact lens development, and it may help reduce both the need for in vivo tests (with ethical and economic advantages) and the gap between the lens design and clinical application. It also allows for an improved understanding of drug transport in the eye.
Collapse
Affiliation(s)
- Nadia Toffoletto
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal.
- Centro de Investigação Interdisciplinar Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511, Caparica, Portugal.
| | - Benilde Saramago
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Ana Paula Serro
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
- Centro de Investigação Interdisciplinar Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511, Caparica, Portugal
| | - Anuj Chauhan
- Chemical and Biological Engineering Department, Colorado School of Mines, Golden, CO, 80401, USA
| |
Collapse
|
14
|
German C, Chen Z, Przekwas A, Walenga R, Babiskin A, Zhao L, Fan J, Tan ML. Computational Model of In Vivo Corneal Pharmacokinetics and Pharmacodynamics of Topically Administered Ophthalmic Drug Products. Pharm Res 2023; 40:961-975. [PMID: 36959411 DOI: 10.1007/s11095-023-03480-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/09/2023] [Indexed: 03/25/2023]
Abstract
INTRODUCTION Although the eye is directly accessible on the surface of the human body, drug delivery can be extremely challenging due to the presence of multiple protective barriers in eye tissues. Researchers have developed complex formulation strategies to overcome these barriers to ophthalmic drug delivery. Current development strategies rely heavily on in vitro experiments and animal testing to predict human pharmacokinetics (PK) and pharmacodynamics (PD). OBJECTIVE The primary objective of the study was to develop a high-fidelity PK/PD model of the anterior eye for topical application of ophthalmic drug products. METHODS Here, we present a physiologically-based in silico approach to predicting PK and PD in rabbits after topical administration of ophthalmic products. A first-principles based approach was used to describe timolol dissolution, transport, and distribution, including consideration of ionized transport, following topical instillation of a timolol suspension. RESULTS Using literature transport and response parameters, the computational model described well the concentration-time and response-time profiles in rabbit. Comparison of validated rabbit model results and extrapolated human model results demonstrate observable differences in the distribution of timolol at multiple time points. CONCLUSION This modeling framework provides a tool for model-based prediction of PK in eye tissues and PD after topical ophthalmic drug administration to the eyes.
Collapse
Affiliation(s)
- Carrie German
- CFD Research Corporation, Computational Biology Division, 6820 Moquin Dr NW, Huntsville, AL, 35806, USA.
| | - Zhijian Chen
- CFD Research Corporation, Computational Biology Division, 6820 Moquin Dr NW, Huntsville, AL, 35806, USA
| | - Andrzej Przekwas
- CFD Research Corporation, Computational Biology Division, 6820 Moquin Dr NW, Huntsville, AL, 35806, USA
| | - Ross Walenga
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Andrew Babiskin
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Liang Zhao
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Jianghong Fan
- Division of Pharmacometrics, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Ming-Liang Tan
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| |
Collapse
|
15
|
Kuepfer L, Fuellen G, Stahnke T. Quantitative systems pharmacology of the eye: Tools and data for ocular QSP. CPT Pharmacometrics Syst Pharmacol 2023; 12:288-299. [PMID: 36708082 PMCID: PMC10014063 DOI: 10.1002/psp4.12918] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 12/21/2022] [Accepted: 01/02/2023] [Indexed: 01/29/2023] Open
Abstract
Good eyesight belongs to the most-valued attributes of health, and diseases of the eye are a significant healthcare burden. Case numbers are expected to further increase in the next decades due to an aging society. The development of drugs in ophthalmology, however, is difficult due to limited accessibility of the eye, in terms of drug administration and in terms of sampling of tissues for drug pharmacokinetics (PKs) and pharmacodynamics (PDs). Ocular quantitative systems pharmacology models provide the opportunity to describe the distribution of drugs in the eye as well as the resulting drug-response in specific segments of the eye. In particular, ocular physiologically-based PK (PBPK) models are necessary to describe drug concentration levels in different regions of the eye. Further, ocular effect models using molecular data from specific cellular systems are needed to develop dose-response correlations. We here describe the current status of PK/PBPK as well as PD models for the eyes and discuss cellular systems, data repositories, as well as animal models in ophthalmology. The application of the various concepts is highlighted for the development of new treatments for postoperative fibrosis after glaucoma surgery.
Collapse
Affiliation(s)
- Lars Kuepfer
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Aging Research (IBIMA), Rostock University Medical Center, Rostock, Germany
| | - Thomas Stahnke
- Institute for ImplantTechnology and Biomaterials e.V., Rostock, Germany.,Department of Ophthalmology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
16
|
Hughes P, Rivers HM, Bantseev V, Yen CW, Mahler HC, Gupta S. Intraocular delivery considerations of ocular biologic products and key preclinical determinations. Expert Opin Drug Deliv 2023; 20:223-240. [PMID: 36632784 DOI: 10.1080/17425247.2023.2166927] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Ophthalmic diseases of the retina are a significant cause of vision loss globally. Despite much progress, there remains an unmet need for durable, long-acting treatment options. While biologic therapies show great promise, they present many challenges, including complexities in biochemical properties, mechanism of action, manufacturing considerations, preclinical evaluation, and delivery mechanism; these are confounded by the unique anatomy and physiology of the eye itself. AREAS COVERED This review describes the current development status of intravitreally administered drugs for the treatment of ophthalmic disease, outlines the range of approaches that can be considered for sustained drug delivery to the eye, and discusses key preclinical considerations for the evaluation of ocular biologics. EXPERT OPINION The required frequency of dosing in the eye results in a great burden on both patients and the health care system, with direct intraocular administration remaining the most reliable and predictable route. Sustained and controlled ophthalmic drug delivery systems will go a long way in reducing this burden. Sustained delivery can directly dose target tissues, improving bioavailability and reducing off-target systemic effects. Maintaining stability and activity of compounds can prevent aggregation and enable extended duration of release, while sustaining dosage and preventing residual polymer after drug depletion.
Collapse
Affiliation(s)
- Patrick Hughes
- Pharmaceutical Development, Visus Therapeutics, Irvine, CA, USA
| | - Hongwen M Rivers
- Biomaterials and Drug Delivery, Medical Aesthetics, AbbVie Inc, North Chicago, IL, USA
| | - Vladimir Bantseev
- Department of Safety Assessment, Genentech, Inc, South San Francisco, CA, USA
| | - Chun-Wan Yen
- Department of Safety Assessment, Genentech, Inc, South San Francisco, CA, USA
| | | | - Swati Gupta
- Non-clinical Development Immunology, AbbVie Inc, North Chicago, IL, USA
| |
Collapse
|
17
|
Alambiaga-Caravaca AM, González Iglesias LG, Rodilla V, Kalia YN, López-Castellano A. Biodistribution of progesterone in the eye after topical ocular administration via drops or inserts. Int J Pharm 2022; 630:122453. [PMID: 36455753 DOI: 10.1016/j.ijpharm.2022.122453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
Progesterone (PG) has been shown to have a slowing effect on photoreceptor cell death in mouse models of retinitis pigmentosa when administered orally. The aim of this study was to investigate whether ophthalmically administered progesterone was able to reach neuroretina and thus, the distribution through ocular tissues of different PG formulations was studied. The effect of different initial PG concentration was also investigated. Different formulations with PG in their composition (drops, a corneal/scleral-insert and scleral-inserts) were prepared and assayed. Using whole porcine eyes, the different formulations were topically administered to the ocular surface. Frozen eyes were dissected, the PG in each tissue was extracted in acetonitrile and the amount of PG quantified by UHPLC-MS/MS. Our results show that after topical administration, PG diffuses from the ocular surface and distributes throughout all tissues of the eye. Lower levels of PG were found in sclera, choroid and neuroretina when PG was applied as drops compared to inserts. Our results also show that an increase in the initial PG concentrations applied, resulted in a statistically significant increase in the amounts of PG in aqueous humour, sclera, choroid and neuroretina.
Collapse
Affiliation(s)
- Adrián M Alambiaga-Caravaca
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115 Valencia, Spain
| | - Laura G González Iglesias
- School of Pharmaceutical Sciences, University of Geneva & University of Lausanne, CMU-1, rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Vicent Rodilla
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115 Valencia, Spain
| | - Yogeshvar N Kalia
- School of Pharmaceutical Sciences, University of Geneva & University of Lausanne, CMU-1, rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Alicia López-Castellano
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115 Valencia, Spain.
| |
Collapse
|
18
|
The Effects of Intravitreal Administration of Antifungal Drugs on the Structure and Mechanical Properties Peripheral Blood Erythrocyte Surface in Rabbits. Int J Mol Sci 2022; 23:ijms231810464. [PMID: 36142376 PMCID: PMC9499686 DOI: 10.3390/ijms231810464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/03/2022] [Accepted: 09/07/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Fungal infections can pose great threat to sight. Immediate treatment is usually required; antifungal agents are widely accepted and are effective in most cases. The present experimental study aims to investigate the probable effects of intravitreal injection of antifungal agents on the structure and mechanical properties of the surface of peripheral blood erythrocytes. Methods: Nine albino New Zealand white rabbits, aged five months old, were chosen for the experiment. Solutions of micafungin, voriconazole, or balanced salt solution (BSS) were injected into the midvitreous. Animals were divided into two experimental groups and one control group. Blood sampling from an intravenous (IV) line was performed after 10 days from the last IV injection. An atomic force microscope (AFM) was used to study the structural and mechanical properties of cell surfaces. Results: The analysis results showed that the parameters of the cytoskeleton’s spatial organization changed insignificantly with the antifungal drug treatment. Conclusions: Our findings suggest that locally administered antifungal drugs can cause significant changes to the structure and frictional properties of the erythrocyte surface. These effects occur in the long-term period after administration of the drugs and represent a potential possibility for violation of blood supply to tissues, and the further development of negative side effects.
Collapse
|
19
|
Marko L, Arto U, Veli-Pekka R. Quantitative pharmacokinetic analyses of anterior and posterior elimination routes of intravitreal anti-VEGF macromolecules using published human and rabbit data. Exp Eye Res 2022; 222:109162. [PMID: 35760120 DOI: 10.1016/j.exer.2022.109162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/17/2022] [Accepted: 06/20/2022] [Indexed: 11/04/2022]
Abstract
The purpose of this study was to evaluate the contribution of the anterior elimination route for four anti-vascular endothelial growth factor (anti-VEGF) macromolecules (aflibercept, bevacizumab, pegaptanib and ranibizumab) after intravitreal injection using published human and rabbit data and three previously described pharmacokinetic (PK) modeling methods. A PubMed search was used to identify published studies with concentration-time data. The data were utilized only if the intravitreally injected drugs were used as plain solutions and several criteria for a well-performed PK study were fulfilled. The three methods to analyze rabbit data were (1) the equation for vitreal elimination half-life based molecular size assuming anterior elimination, (2) Maurice equation and plot for the ratio of aqueous humor (AH) to vitreal concentration assuming anterior elimination, and (3) the equation for amount of macromolecule eliminated anteriorly based on the area under the curve in AH. The first and third methods were used for human data. In the second and third methods, AH flow rate is a key model parameter, and it was varied between 2 and 3 μl/min. The methods were applied to data from 9 rabbit studies (1 for aflibercept, 5 for bevacizumab, and 3 for ranibizumab) and 5 human studies (1 for aflibercept, 3 for bevacizumab, and 1 for ranibizumab). Experimental half-lives of anti-VEGF macromolecules in both vitreous and aqueous humor were close to those calculated with the equations for vitreal elimination half-life in humans and rabbits. Rabbit data analyzed with Maurice plot indicated that the contribution of anterior elimination was usually at least 75%. In most human and rabbit studies, the calculated percentage of anterior elimination was at least 51%. Variability between studies was extensive for bevacizumab and ranibizumab. The results suggest that the anterior elimination route dominates after intravitreal injection of anti-VEGF macromolecules. However, the clinical data are sparse and variability is extensive, the latter emphasizing the need of proper experimental design.
Collapse
Affiliation(s)
- Lamminsalo Marko
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Urtti Arto
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland; Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, St. Petersburg, Russian Federation
| | - Ranta Veli-Pekka
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
20
|
Yang Y, Zhang X. Integration of Engineered Delivery with the Pharmacokinetics of Medical Candidates via Physiology-Based Pharmacokinetics. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2486:57-69. [PMID: 35437718 DOI: 10.1007/978-1-0716-2265-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is a mechanistic computational model that can be used to predict a drug product's ADME (absorption, distribution, metabolism, and excretion) and pharmacokinetics (PK). In recent years, PBPK modeling and simulation has been used increasingly to address many biopharmaceutics and clinical pharmacology questions, such as the effect of formulations, intrinsic factors (age, organ dysfunction, etc.), and extrinsic factors (comedications, food) on the PK of an investigational drug product. In this chapter, we will briefly introduce various PBPK models for ADME prediction and general procedures for PBPK modeling and simulations. The readers are encouraged to read updated literature on new applications of PBPK modeling and simulation which is still an emerging area in pharmaceutical development.
Collapse
Affiliation(s)
- Yuching Yang
- Division of Pharmacometrics, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xinyuan Zhang
- Division of Pharmacometrics, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA.
| |
Collapse
|
21
|
Kelley RF, Tesar DB, Wang Y, Agard NJ, Holder PG, Chan J, Comps-Agrar L, Horvath J, Horvath JD, Crowell SR. Generation of a Porcine Antibody Fab Fragment Using Protein Engineering to Facilitate the Evaluation of Ocular Sustained Delivery Technology. Mol Pharm 2022; 19:1540-1547. [PMID: 35393854 DOI: 10.1021/acs.molpharmaceut.2c00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Treatment of age-related macular degeneration (AMD) with anti-vascular endothelial growth factor (VEGF) biologic agents has been shown to restore and maintain visual acuity for many patients afflicted with wet AMD. These agents are usually administered via intravitreal injection at a dosing interval of 4-8 weeks. Employment of long-acting delivery (LAD) technologies could improve the therapeutic outcome, ensure timely treatment, and reduce burden on patients, caregivers, and the health care system. Development of LAD approaches requires thorough testing in pre-clinical species; however, therapeutic proteins of human origin may not be well tolerated during testing in non-human species due to immunogenicity. Here, we have engineered a surrogate porcine antibody Fab fragment (pigG6.31) from a human antibody for testing ocular LAD technologies in a porcine model. The engineered Fab retains the VEGF-A-binding and inhibition properties of the parental human Fab and has stability properties suitable for LAD evaluation. Upon intravitreal injection in minipigs, pigG6.31 showed first-order clearance from the ocular compartments with vitreal elimination rates consistent with other molecules of this size. Application of the surrogate molecule in an in vivo evaluation in minipigs of a prototype of the port delivery (PD) platform indicated continuous ocular delivery from the implant, with release kinetics consistent with both the results from in vitro release studies and the efficacy observed in human clinical studies of the PD system with ranibizumab (PDS). Anti-drug antibodies in the serum against pigG6.31 were not detected over exposure durations up to 16 weeks, suggesting that this molecule has low porcine immunogenicity.
Collapse
Affiliation(s)
- Robert F Kelley
- Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Devin B Tesar
- Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yue Wang
- Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Nicholas J Agard
- Antibody Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Patrick G Holder
- Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joyce Chan
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Laetitia Comps-Agrar
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Judit Horvath
- Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joshua D Horvath
- Device Development, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Susan R Crowell
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics Department, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
22
|
Chaffiol A, Provansal M, Joffrois C, Blaize K, Labernede G, Goulet R, Burban E, Brazhnikova E, Duebel J, Pouget P, Sahel JA, Picaud S, Arcizet F, Gauvain G. In vivo optogenetic stimulation of the primate retina activates the visual cortex after long-term transduction. Mol Ther Methods Clin Dev 2022; 24:1-10. [PMID: 34977267 PMCID: PMC8671818 DOI: 10.1016/j.omtm.2021.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/19/2021] [Indexed: 11/26/2022]
Abstract
Over the last 15 years, optogenetics has changed fundamental research in neuroscience and is now reaching toward therapeutic applications. Vision restoration strategies using optogenetics are now at the forefront of these new clinical opportunities. But applications to human patients suffering from retinal diseases leading to blindness raise important concerns on the long-term functional expression of optogenes and the efficient signal transmission to higher visual centers. Here, we demonstrate in non-human primates continued expression and functionality at the retina level ∼20 months after delivery of our construct. We also performed in vivo recordings of visually evoked potentials in the primary visual cortex of anesthetized animals. Using synaptic blockers, we isolated the in vivo cortical activation resulting from the direct optogenetic stimulation of primate retina. In conclusion, our work indicates long-term transgene expression and transmission of the signal generated in the macaque retina to the visual cortex, two important features for future clinical applications.
Collapse
Affiliation(s)
- Antoine Chaffiol
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Matthieu Provansal
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Corentin Joffrois
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Kévin Blaize
- Institut de Neurosciences de la Timone, UMR 7289 Centre National de la Recherche Scientifique and Aix-Marseille Université, 13385 Marseille Cedex 05, France
| | - Guillaume Labernede
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Ruben Goulet
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Emma Burban
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Elena Brazhnikova
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Jens Duebel
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| | - Pierre Pouget
- INSERM 1127, CNRS 7225, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, 75013 Paris, France
| | - José Alain Sahel
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Serge Picaud
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Fabrice Arcizet
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| | - Gregory Gauvain
- Sorbonne Université, Inserm, CNRS, Institut de la Vision, 75012 Paris, France
| |
Collapse
|
23
|
Khoobyar A, Penkova AN, Humayun MS, Sadhal SS. Mathematical Model of Macromolecular Drug Transport in a Partially Liquefied Vitreous Humor. JOURNAL OF HEAT TRANSFER 2022; 144:031208. [PMID: 35833154 PMCID: PMC8823200 DOI: 10.1115/1.4053197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/01/2021] [Indexed: 05/30/2023]
Abstract
The purpose of this study is to investigate the effect of partial liquefaction (due to ageing) of the vitreous humor on the transport of ocular drugs. In our model, the gel part of the vitreous is treated as a Darcy-type porous medium. A spherical region within the porous part of vitreous is in a liquid state which, for computational purposes, is also treated as a porous medium but with a much higher permeability. Using the finite element method, a time-dependent, three-dimensional model has been developed to computationally simulate (using the Petrov-Galerkin method) the transport of intravitreally injected macromolecules where both convection and diffusion are present. From a fluid physics and transport phenomena perspective, the results show many interesting features. For pressure-driven flow across the vitreous, the flow streamlines converge into the liquefied region as the flow seeks the fastest path of travel. Furthermore, as expected, with increased level of liquefaction, the overall flow rate increases for a given pressure drop. We have quantified this effect for various geometrical considerations. The flow convergence into the liquefied region has important implication for convective transport. One effect is the clear diversion of the drug as it reaches the liquefied region. In some instances, the entry point of the drug in the retinal region gets slightly shifted due to liquefaction. While the model has many approximations and assumptions, the focus is illustrating the effect of liquefaction as one of the building blocks toward a fully comprehensive model.
Collapse
Affiliation(s)
- Anahid Khoobyar
- Department of Aerospace and Mechanical Engineering, University of Southern California, USC Viterbi School of Engineering, Los Angeles, CA 90089-1453
| | - Anita N. Penkova
- Department of Aerospace and Mechanical Engineering, USC Viterbi School of Engineering, Los Angeles, CA 90089-1453; Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027
| | - Mark S. Humayun
- Cornelius Pings Professor of Biomedical Sciences, Professor of Ophthalmology, Biomedical Engineering, and Integrative Anatomical Sciences, Director, USC Ginsburg Institute for Biomedical Therapeutics, Co-Director USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033-4682
| | - Satwindar Singh Sadhal
- Department of Aerospace and Mechanical Engineering, University of Southern California, USC Viterbi School of Engineering, Los Angeles, CA 90089-1453; Children's Hospital Los Angeles, Saban Research Institute, Los Angeles, CA 90027; Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033-4682
| |
Collapse
|
24
|
Li H, Zhu X, Wang M, Zhao D, Li H, Yan J. Drug sustained release from degradable drug-loaded in-situ hydrogels in the posterior eye: a mechanistic model and analytical method. J Biomech 2022; 136:111052. [DOI: 10.1016/j.jbiomech.2022.111052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022]
|
25
|
Singh V, Garg A, Dewangan HK. Recent Advances in Drug Design and Delivery Across Biological Barriers using Computational Models. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819999220204110306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
The systemic delivery of pharmacological substances generally exhibits several significant limitations associated with the bio-distribution of active drugs in the body. As per consequence, human body’s defense mechanisms become impediments to drug delivery. Various technologies to overcome these limitations have been evolved including computational approaches and advanced drug delivery. As the body of human has evolved to defend itself from hostile biological as well as chemical invaders, along with that these biological barriers such as ocular barriers, blood-brain barriers, intestinal and skin barriers also limit the passage of drugs across desired sites. Therefore, efficient delivery remains an utmost challenge for researchers and scientists. The present review focuses on the techniques to deliver the drugs with efficient therapeutic efficacy at the targeted sites. This review article considered the insights into main biological barriers along with the application of computational or numerical methods dealing with different barriers by determining the drug flow, temperature and various other parameters. It also summarizes the advanced implantable drug delivery system to circumvent the inherent resistance showed by these biological barriers and in turn to improve the drug delivery.
Collapse
Affiliation(s)
- Vanshita Singh
- Institute of Pharmaceutical Research, GLA University Mathura, NH-2 Delhi Mathura Road, PO-Chaumuhan, Mathura, UttarPradesh, India 281406
| | - Akash Garg
- Institute of Pharmaceutical Research, GLA University Mathura, NH-2 Delhi Mathura Road, PO-Chaumuhan, Mathura, UttarPradesh, India 281406
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University NH-95, Chandigarh Ludhiyana Highway, Mohali Punjab, India
| |
Collapse
|
26
|
Chan D, Won GJ, Read AT, Ethier CR, Thackaberry E, Crowell SR, Booler H, Bantseev V, Sivak JM. Application of an organotypic ocular perfusion model to assess intravitreal drug distribution in human and animal eyes. J R Soc Interface 2022; 19:20210734. [PMID: 35078337 PMCID: PMC8790337 DOI: 10.1098/rsif.2021.0734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Intravitreal (ITV) drug delivery is a new cornerstone for retinal therapeutics. Yet, predicting the disposition of formulations in the human eye remains a major translational hurdle. A prominent, but poorly understood, issue in pre-clinical ITV toxicity studies is unintended particle movements to the anterior chamber (AC). These particles can accumulate in the AC to dangerously raise intraocular pressure. Yet, anatomical differences, and the inability to obtain equivalent human data, make investigating this issue extremely challenging. We have developed an organotypic perfusion strategy to re-establish intraocular fluid flow, while maintaining homeostatic pressure and pH. Here, we used this approach with suitably sized microbeads to profile anterior and posterior ITV particle movements in live versus perfused porcine eyes, and in human donor eyes. Small-molecule suspensions were then tested with the system after exhibiting differing behaviours in vivo. Aggregate particle size is supported as an important determinant of particle movements in the human eye, and we note these data are consistent with a poroelastic model of bidirectional vitreous transport. Together, this approach uses ocular fluid dynamics to permit, to our knowledge, the first direct comparisons between particle behaviours from human ITV injections and animal models, with potential to speed pre-clinical development of retinal therapeutics.
Collapse
Affiliation(s)
- D. Chan
- Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - G. J. Won
- Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - A. T. Read
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, USA
| | - C. R. Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, USA
| | - E. Thackaberry
- Safety Assessment, Genentech Inc., San Francisco, CA, USA
| | - S. R. Crowell
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics (PTPK) Genentech Inc., San Francisco, CA, USA
| | - H. Booler
- Safety Assessment, Genentech Inc., San Francisco, CA, USA
| | - V. Bantseev
- Safety Assessment, Genentech Inc., San Francisco, CA, USA
| | - J. M. Sivak
- Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada,Department of Ophthalmology and Vision Science, University of Toronto, Toronto, Ontario, Canada,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Ocular Fluid Mechanics and Drug Delivery: A Review of Mathematical and Computational Models. Pharm Res 2021; 38:2003-2033. [PMID: 34936067 DOI: 10.1007/s11095-021-03141-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Abstract
The human eye is a complex biomechanical structure with a range of biomechanical processes involved in various physiological as well as pathological conditions. Fluid flow inside different domains of the eye is one of the most significant biomechanical processes that tend to perform a wide variety of functions and when combined with other biophysical processes play a crucial role in ocular drug delivery. However, it is quite difficult to comprehend the effect of these processes on drug transport and associated treatment experimentally because of ethical constraints and economic feasibility. Computational modeling on the other hand is an excellent means to understand the associated complexity between these aforementioned processes and drug delivery. A wide range of computational models specific to different types of fluids present in different domains of the eye as well as varying drug delivery modes has been established to understand the fluid flow behavior and drug transport phenomenon in an insilico manner. These computational models have been used as a non-invasive tool to aid ophthalmologists in identifying the challenges associated with a particular drug delivery mode while treating particular eye diseases and to advance the understanding of the biomechanical behavior of the eye. In this regard, the author attempts to summarize the existing computational and mathematical approaches proposed in the last two decades for understanding the fluid mechanics and drug transport associated with different domains of the eye, together with their application to modify the existing treatment processes.
Collapse
|
28
|
Kicková E, Sadeghi A, Puranen J, Tavakoli S, Sen M, Ranta VP, Arango-Gonzalez B, Bolz S, Ueffing M, Salmaso S, Caliceti P, Toropainen E, Ruponen M, Urtti A. Pharmacokinetics of Pullulan-Dexamethasone Conjugates in Retinal Drug Delivery. Pharmaceutics 2021; 14:pharmaceutics14010012. [PMID: 35056906 PMCID: PMC8779473 DOI: 10.3390/pharmaceutics14010012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 12/11/2022] Open
Abstract
The treatment of retinal diseases by intravitreal injections requires frequent administration unless drug delivery systems with long retention and controlled release are used. In this work, we focused on pullulan (≈67 kDa) conjugates of dexamethasone as therapeutic systems for intravitreal administration. The pullulan-dexamethasone conjugates self-assemble into negatively charged nanoparticles (average size 326 ± 29 nm). Intravitreal injections of pullulan and pullulan-dexamethasone were safe in mouse, rat and rabbit eyes. Fluorescently labeled pullulan particles showed prolonged retention in the vitreous and they were almost completely eliminated via aqueous humor outflow. Pullulan conjugates also distributed to the retina via Müller glial cells when tested in ex vivo retina explants and in vivo. Pharmacokinetic simulations showed that pullulan-dexamethasone conjugates may release free and active dexamethasone in the vitreous humor for over 16 days, even though a large fraction of dexamethasone may be eliminated from the eye as bound pullulan-dexamethasone. We conclude that pullulan based drug conjugates are promising intravitreal drug delivery systems as they may reduce injection frequency and deliver drugs into the retinal cells.
Collapse
Affiliation(s)
- Eva Kicková
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (E.K.); (S.S.); (P.C.)
| | - Amir Sadeghi
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland; (A.S.); (J.P.); (V.-P.R.); (E.T.); (M.R.)
| | - Jooseppi Puranen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland; (A.S.); (J.P.); (V.-P.R.); (E.T.); (M.R.)
| | - Shirin Tavakoli
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00710 Helsinki, Finland;
| | - Merve Sen
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, D-72076 Tübingen, Germany; (M.S.); (B.A.-G.); (S.B.); (M.U.)
| | - Veli-Pekka Ranta
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland; (A.S.); (J.P.); (V.-P.R.); (E.T.); (M.R.)
| | - Blanca Arango-Gonzalez
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, D-72076 Tübingen, Germany; (M.S.); (B.A.-G.); (S.B.); (M.U.)
| | - Sylvia Bolz
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, D-72076 Tübingen, Germany; (M.S.); (B.A.-G.); (S.B.); (M.U.)
| | - Marius Ueffing
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, D-72076 Tübingen, Germany; (M.S.); (B.A.-G.); (S.B.); (M.U.)
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (E.K.); (S.S.); (P.C.)
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (E.K.); (S.S.); (P.C.)
| | - Elisa Toropainen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland; (A.S.); (J.P.); (V.-P.R.); (E.T.); (M.R.)
| | - Marika Ruponen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland; (A.S.); (J.P.); (V.-P.R.); (E.T.); (M.R.)
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland; (A.S.); (J.P.); (V.-P.R.); (E.T.); (M.R.)
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00710 Helsinki, Finland;
- Institute of Chemistry, St. Petersburg State University, Petergof, Universitetskii pr. 26, 198504 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
29
|
Impact of silicone oil tamponade on intravitreally injected vancomycin pharmacokinetics in cynomolgus monkey eyes. Int J Pharm 2021; 609:121185. [PMID: 34655708 DOI: 10.1016/j.ijpharm.2021.121185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/26/2021] [Accepted: 10/10/2021] [Indexed: 11/23/2022]
Abstract
Intravitreal injections of vancomycin (VCM) and ceftazidime (CAZ) are commonly used to treat infectious endophthalmitis. When patient cases require retinal detachment with silicone oil (SO) tamponade, the antibiotic doses are empirically reduced to 25 %. Currently, there is no scientific evidence for these empirical dose reductions. The purpose of the present study is to determine the quantitative impact that SO tamponades have on intraocular VCM pharmacokinetics. Because of high invasiveness of frequent sampling of intraocular VCM concentrations in human, this pharmacokinetic study was performed in cynomolgus monkey's eyes. Population pharmacokinetic modeling and simulation were performed using 75 different intraocular VCM concentrations obtained from 8 male cynomolgus monkeys. A one-compartment model with a first-order diffusion rate was used as a structural pharmacokinetic model. From the covariate analysis, SO tamponade significantly decreased the volume of distribution while pars plana vitrectomy with lensectomy (PPV) significantly increased the clearance and diffusion rate constants. From the Monte Carlo simulation (n = 1,000), the median time above minimum inhibitory concentration (T>MIC, a therapeutic effect index) durations of SO and normal eyes at clinical doses of 1,000 µg were 2.6 and 11.0 days, respectively. Using intravitreal injections of VCM with SO tamponade or PPV may reduce the therapeutic effect.
Collapse
|
30
|
Litvin G, Klein I, Litvin Y, Klaiman G, Nyska A. CorNeat KPro: Ocular Implantation Study in Rabbits. Cornea 2021; 40:1165-1174. [PMID: 34351873 PMCID: PMC8330828 DOI: 10.1097/ico.0000000000002798] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 04/26/2021] [Accepted: 05/06/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE The purpose of this study was to evaluate surgical feasibility and long-term integration of the CorNeat Keratoprosthesis (KPro), a novel synthetic cornea, in rabbits. METHODS The CorNeat KPro is a synthetic corneal implant designed to treat corneal blindness by using a polymeric scaffold for biointegration, consequently assimilating synthetic optics within ocular tissues. Eight New Zealand White rabbits were implanted unilaterally with the CorNeat KPro and observed for 6 months. Animals were regularly monitored by a certified ophthalmologist using slit-lamp biomicroscopy. One animal developed postoperative endophthalmitis and was removed from the study 7 weeks postsurgery. At termination, eyes were enucleated and evaluated histologically to assess local tissue integration and inflammatory response. RESULTS The surgical procedure was found feasible. The CorNeat KPro integrated into all operated eyes, resulting in a retention rate of 87.5% at the conclusion of the 6-month follow-up period. We observed minimal-to-mild conjunctival and iridial congestion and did not find additional inflammatory indicators, such as anterior chamber fibrin, flare, or cells. The optical element of the device remained clear with zero incidence of retroprosthetic membrane formation. Histopathological evaluation revealed comparable tissue and cellular reaction in all eyes, consisting of the presence of fibroblasts and associated collagen fibrils within the device's skirt component. Some eyes showed a mild foreign body reaction surrounding the skirt. CONCLUSIONS Clinical and histological findings indicate the integration of the implanted device into the surrounding tissue, evident by the retention rate and the diffuse infiltration of fibroblasts with collagen deposition among the device's fibrils. These data hold promise for clinical application in humans.
Collapse
Affiliation(s)
| | - Ido Klein
- CorNeat Vision Ltd, Raanana, Israel;
| | - Yoav Litvin
- Independent Scientific Consultant, Bellingham, WA
| | - Guy Klaiman
- Envigo CRS (Israel), Ness Ziona, Israel; and
| | - Abraham Nyska
- Sackler School of Medicine, Consultant in Toxicologic Pathology, Timrat and Tel Aviv University, Israel.
| |
Collapse
|
31
|
Adrianto MF, Annuryanti F, Wilson CG, Sheshala R, Thakur RRS. In vitro dissolution testing models of ocular implants for posterior segment drug delivery. Drug Deliv Transl Res 2021; 12:1355-1375. [PMID: 34382178 PMCID: PMC9061687 DOI: 10.1007/s13346-021-01043-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 12/19/2022]
Abstract
The delivery of drugs to the posterior segment of the eye remains a tremendously difficult task. Prolonged treatment in conventional intravitreal therapy requires injections that are administered frequently due to the rapid clearance of the drug molecules. As an alternative, intraocular implants can offer drug release for long-term therapy. However, one of the several challenges in developing intraocular implants is selecting an appropriate in vitro dissolution testing model. In order to determine the efficacy of ocular implants in drug release, multiple in vitro test models were emerging. While these in vitro models may be used to analyse drug release profiles, the findings may not predict in vivo retinal drug exposure as this is influenced by metabolic and physiological factors. This review considers various types of in vitro test methods used to test drug release of ocular implants. Importantly, it discusses the challenges and factors that must be considered in the development and testing of the implants in an in vitro setup.
Collapse
Affiliation(s)
- Muhammad Faris Adrianto
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Airlangga, Surabaya, East Java, 60115, Indonesia
| | - Febri Annuryanti
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Airlangga, Surabaya, East Java, 60115, Indonesia
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland
| | - Ravi Sheshala
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300, Bandar Puncak Alam, Kuala Selangor, Malaysia
| | - Raghu Raj Singh Thakur
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
32
|
Rimpelä AK, Cui Y, Sauer A. Mechanistic Model for the Prediction of Small-Molecule Vitreal Clearance Combining Diffusion-Limited and Permeability-Limited Clearance. Mol Pharm 2021; 18:2703-2713. [PMID: 34151575 DOI: 10.1021/acs.molpharmaceut.1c00236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The discovery of new small-molecule drugs for intravitreal administration would benefit from simple models to predict vitreal clearance (CL). The current models available have limitations in their applicability to small-molecule drugs and translatability to humans. We developed a mechanistic model combining the diffusion rate of the molecule in the vitreous and permeability across posterior segment tissues and applied it to 30 small molecules with observed CL available mostly from literature. We used Caco-2 permeability as a surrogate for ocular tissue permeability. The model predicted rabbit vitreal CL well, with 80% of the predictions being within a 2-fold range of the observed CL. For an accurate prediction, it was crucial to consider the anterior diffusion CL from the vitreous to the aqueous and a limiting diffusion CL for the whole eye. We observed no major differences in model accuracy when using literature permeability values from retinal pigment epithelial cell models. Importantly, by adopting the specific dimensions of the human eye, the model was able to accurately predict vitreal CL of four compounds for which human vitreal CL data are available. In summary, this mechanistic model enables a simple, accurate, and translatable estimation of small-molecule vitreal CL.
Collapse
Affiliation(s)
- Anna-Kaisa Rimpelä
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Yunhai Cui
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Achim Sauer
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| |
Collapse
|
33
|
Multi-channel transorbital electrical stimulation for effective stimulation of posterior retina. Sci Rep 2021; 11:9745. [PMID: 33963229 PMCID: PMC8105361 DOI: 10.1038/s41598-021-89243-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/23/2021] [Indexed: 02/03/2023] Open
Abstract
Transorbital electrical stimulation (tES) has been studied as a new noninvasive method for treating intractable eye diseases by delivering weak electrical current to the eye through a pair of electrodes attached to the skin around the eye. Studies have reported that the therapeutic effect of tES is determined by the effective stimulation of retinal cells that are densely distributed in the posterior part of the retina. However, in conventional tES with a pair of electrodes, a greater portion of the electric field is delivered to the anterior part of the retina. In this study, to address this issue, a new electrode montage with multiple electrodes was proposed for the effective delivery of electric fields to the posterior retina. Electric field analysis based on the finite element method was performed with a realistic human head model, and optimal injection currents were determined using constrained convex optimization. The resultant electric field distributions showed that the proposed multi-channel tES enables a more effective stimulation of the posterior retina than the conventional tES with a pair of electrodes.
Collapse
|
34
|
Kompella UB, Hartman RR, Patil MA. Extraocular, periocular, and intraocular routes for sustained drug delivery for glaucoma. Prog Retin Eye Res 2021; 82:100901. [PMID: 32891866 PMCID: PMC8317199 DOI: 10.1016/j.preteyeres.2020.100901] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 08/22/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
Although once daily anti-glaucoma drug therapy is a current clinical reality, most therapies require multiple dosing and there is an unmet need to develop convenient, safe, and effective sustained release drug delivery systems for long-term treatment to improve patient adherence and outcomes. One of the first sustained release drug delivery systems was approved for the reduction of intraocular pressure in glaucoma patients. It is a polymeric reservoir-type insert delivery system, Ocusert™, placed under the eyelid and on the ocular surface for zero-order drug release over one week. The insert, marketed in two strengths, released pilocarpine on the eye surface. While many clinicians appreciated this drug product, it was eventually discontinued. No similar sustained release non-invasive drug delivery system has made it to the market to date for treating glaucoma. Drug delivery systems under development include punctal plugs, ring-type systems, contact lenses, implants, microspheres, nanospheres, gels, and other depot systems placed in the extraocular, periocular, or intraocular regions including intracameral, supraciliary, and intravitreal spaces. This article discusses the advantages and disadvantages of the various routes of administration and delivery systems for sustained glaucoma therapy. It also provides the reader with some examples and discussion of drug delivery systems that could potentially be applied for glaucoma treatment. Interestingly, one intracamerally injected implant, Durysta™, was approved recently for sustained intraocular pressure reduction. However, long-term acceptance of such devices has yet to be established. The ultimate success of the delivery system will depend on efficacy relative to eye drop dosing, safety, reimbursement options, and patient acceptance. Cautious development efforts are warranted considering prior failed approaches for sustained glaucoma drug delivery. Neuroprotective approaches for glaucoma therapy including cell, gene, protein, and drug-combination therapies, mostly administered intravitreally, are also rapidly progressing towards assessment in humans.
Collapse
Affiliation(s)
- Uday B Kompella
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Rachel R Hartman
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Madhoosudan A Patil
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
35
|
Kiss S, Oresic Bender K, Grishanin RN, Hanna KM, Nieves JD, Sharma P, Nguyen AT, Rosario RJ, Greengard JS, Gelfman CM, Gasmi M. Long-Term Safety Evaluation of Continuous Intraocular Delivery of Aflibercept by the Intravitreal Gene Therapy Candidate ADVM-022 in Nonhuman Primates. Transl Vis Sci Technol 2021; 10:34. [PMID: 33532145 PMCID: PMC7846953 DOI: 10.1167/tvst.10.1.34] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/06/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose To evaluate the long-term safety of vascular endothelial growth factor (VEGF) suppression with sustained aflibercept expression after a single intravitreal injection (IVI) of ADVM-022, an anti-VEGF gene therapy, in non-human primates (NHPs). Methods Non-human primates received bilateral IVI of ADVM-022, a gene therapy vector encoding aflibercept, a standard of care for the treatment of VEGF-based retinal disease. Aflibercept levels from ocular fluids and tissues were measured. Ocular inflammation was assessed by slit lamp biomicroscopy and fundoscopy. The integrity of the retinal structure was analyzed by optical coherence tomography and blue light fundus autofluorescence and electroretinography was performed to determine retinal function. Histologic evaluation of the retina was performed at the longest time point measured (2.5 years after injection). Results Sustained expression of aflibercept was noted out to the last time point evaluated. Mild to moderate inflammatory responses were observed, which trended toward spontaneous resolution without anti-inflammatory treatment. No abnormalities in retinal structure or function were observed, as measured by optical coherence tomography and electroretinography, respectively. RPE integrity was maintained throughout the study; no histologic abnormalities were observed 2.5 years after ADVM-022 IVI. Conclusions In non-human primates, long-term, sustained aflibercept expression and the resulting continuous VEGF suppression by a single IVI of ADVM-022, appears to be safe, with no measurable adverse effects on normal retinal structure and function evaluated out to 2.5 years. Translational Relevance Together with the results from previous ADVM-022 preclinical studies, these data support the evaluation of this gene therapy candidate in clinical trials as a potential durable treatment for various VEGF-mediated ophthalmic disorders.
Collapse
Affiliation(s)
- Szilárd Kiss
- Weill Cornell Medical College, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | - Mehdi Gasmi
- Adverum Biotechnologies, Inc. Redwood City, CA USA
| |
Collapse
|
36
|
Abootorabi S, Tripathi A, Yu HW, Dávila LP. Computational modeling of intraocular drug delivery supplied by porous implants. Drug Deliv Transl Res 2021; 11:2134-2143. [PMID: 33432523 DOI: 10.1007/s13346-020-00878-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 11/27/2022]
Abstract
New and efficient drug delivery to the posterior part of the eye is a growing health necessity worldwide. Current treatment of eye diseases, such as age-related macular degeneration (AMD), relies on repeated intravitreal injections of drug-containing solutions. Such a drug delivery has major drawbacks including short drug life, significant medical service, and high medical cost. In this study, we explored a new approach to controlled drug delivery by introducing unique porous implants. Our computational modeling contained key physiological and anatomical traits. Incompressible flow in a porous media field, including the sclera, choroid, and retina layers, is governed by Darcy law and the time evolution of the drug concentration was solved via three convection-diffusion equations in the three layers, respectively. The computational model was validated by established results from independent studies and experimental data. Simulations of the IgG1 Fab drug delivery to the posterior eye were performed to evaluate the effectiveness of the porous implants for controlled delivery. Overall, our results indicate that drug therapeutic levels in the posterior eye sustain for eight weeks similarly to those using intravitreal injection. We first evaluated the effects of the porous implants on the drug delivery in the posterior layers. Subsequent simulations were carried out with varying porosity values in a porous episcleral implant. We found that the time evolution of drug concentration is distinctively correlated to drug source location and pore size. A correlation between porosity and fluid properties for selected porous implants was revealed for the first time in this study.
Collapse
Affiliation(s)
- Seyedalireza Abootorabi
- Department of Mechanical and Energy Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University, 723 West Michigan Street, Indianapolis, IN, 46202, USA
| | - Abhimanyu Tripathi
- Department of Materials Science and Engineering, School of Engineering, University of California Merced, 5200 N. Lake Road, Merced, CA, 95343, USA
| | - Huidan Whitney Yu
- Department of Mechanical and Energy Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University, 723 West Michigan Street, Indianapolis, IN, 46202, USA
| | - Lilian P Dávila
- Department of Materials Science and Engineering, School of Engineering, University of California Merced, 5200 N. Lake Road, Merced, CA, 95343, USA.
| |
Collapse
|
37
|
Bhandari A, Bansal A, Sinha N. Effect of aging on heat transfer, fluid flow and drug transport in anterior human eye: A computational study. J Control Release 2020; 328:286-303. [PMID: 32861760 DOI: 10.1016/j.jconrel.2020.08.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/07/2020] [Accepted: 08/22/2020] [Indexed: 12/15/2022]
Abstract
There are a lot of geometrical and morphological changes that happen in the human eye with age. Primary open-angle glaucoma, which is caused by the increase in intraocular pressure inside the anterior chamber of the eye is also associated with the physiological aging of the eye. Therefore, it is crucial to understand the effects of aging on drug delivery in the human eye when applied topically. Consequently, a numerical model of topical drug delivery for an aging human eye has been developed using commercial software COMSOL Multiphysics in the current study. Three different age groups (young, middle and old) have been considered and the changes in geometrical and tissue properties of different domains of the eye with age have been included in the numerical model. The effect of aging on heat transfer, aqueous humor flow, intraocular pressure and drug concentration in different domains and orientations of the eye have been investigated. Additionally, an attempt has been made to predict the best class of anti-glaucomatic treatment in silico that should be preferred to treat primary open-angle glaucoma effectively. Results illustrate that there is a decrease in the average corneal temperature and an increase in the temperature deviation across the cornea with age. Further, there is a decrease in the aqueous humor flow magnitude in the anterior chamber of the eye and an increase in intraocular pressure in the anterior chamber of older age groups, which leads to primary open-angle glaucoma. The reduced aqueous humor flow leads to increased drug concentration in the anterior chamber as well as iris and reduced drug concentration in the trabecular mesh of the older age groups, thereby affecting the treatment efficacy. Additionally, our simulated results demonstrate that anti-glaucomatic treatments should be more focused on treating the trabecular mesh rather than the ciliary body of the eye.
Collapse
Affiliation(s)
- Ajay Bhandari
- Department of Mechanical Engineering, Indian Institute of Technology, Kanpur 208016, India; Department of Mechanical Engineering, Indian Institute of Technology (Indian School of Mines), Dhanbad, Jharkhand 826004, India
| | - Ankit Bansal
- Department of Mechanical and Industrial Engineering, Indian Institute of Technology, Roorkee 247677, India
| | - Niraj Sinha
- Department of Mechanical Engineering, Indian Institute of Technology, Kanpur 208016, India.
| |
Collapse
|
38
|
Huang X, Chau Y. Enhanced Delivery of siRNA to Retinal Ganglion Cells by Intravitreal Lipid Nanoparticles of Positive Charge. Mol Pharm 2020; 18:377-385. [PMID: 33295773 DOI: 10.1021/acs.molpharmaceut.0c00992] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
RNAi therapy has been developed and explored for treating retinal conditions since last decades. The progression of retinal diseases including the age-related macular degeneration and glaucoma is associated with the malfunction of specific retinal cells. Therefore, to deliver therapeutic RNAi to selective retinal tissues with desired gene downregulation is crucial for the treatment of retinal diseases via RNAi therapy. Lipid-based nanoparticles are potent delivery vectors for RNAi therapeutics to achieve high gene silencing efficiency. The surface charge has been demonstrated to affect the intraocular behaviors and retinal distribution of intravitreally administered lipid nanoparticles (LNPs), which could subsequently affect the gene knockdown efficiency in specific retinal layers. Here, we evaluated three charged LNPs for their ability to deliver siRNA and facilitate gene downregulation both in vitro and in vivo. LNPs with different surface charges ranging from neutral to positive (5-34 mV) were successfully formulated. All types of charged LNPs managed gene knockdown in both mammalian cell line and primary neurons. At 48 h post intravitreal injection, neutral LNPs (6.2 mV) and mildly positive LNPs (15.9 mV) mediated limited retinal gene suppression (<10%) and the more positive LNPs (31.2 mV) led to ∼25% gene suppression in the retinal ganglion cell (RGC) layer. No gene silencing in the retinal pigmented epithelium layer was facilitated by any LNPs independent of the charges. In summary, this study has shown that positive LNPs with an optimized charge managed specific gene downregulation in the RGC layer. These RNAi carriers hold potential for the treatment of RGC-associated retinal diseases.
Collapse
Affiliation(s)
- Xiaonan Huang
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clearwater Bay, Kowloon 999077, Hong Kong, China
| | - Ying Chau
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clearwater Bay, Kowloon 999077, Hong Kong, China
| |
Collapse
|
39
|
Bhattacharya M, Sadeghi A, Sarkhel S, Hagström M, Bahrpeyma S, Toropainen E, Auriola S, Urtti A. Release of functional dexamethasone by intracellular enzymes: A modular peptide-based strategy for ocular drug delivery. J Control Release 2020; 327:584-594. [DOI: 10.1016/j.jconrel.2020.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 12/19/2022]
|
40
|
Lamminsalo M, Karvinen T, Subrizi A, Urtti A, Ranta VP. Extended Pharmacokinetic Model of the Intravitreal Injections of Macromolecules in Rabbits. Part 2: Parameter Estimation Based on Concentration Dynamics in the Vitreous, Retina, and Aqueous Humor. Pharm Res 2020; 37:226. [PMID: 33094404 PMCID: PMC7581578 DOI: 10.1007/s11095-020-02946-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/05/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE To estimate the diffusion coefficients of an IgG antibody (150 kDa) and its antigen-binding fragment (Fab; 50 kDa) in the neural retina (Dret) and the combined retinal pigment epithelium-choroid (DRPE-cho) with a 3-dimensional (3D) ocular pharmacokinetic (PK) model of the rabbit eye. METHODS Vitreous, retina, and aqueous humor concentrations of IgG and Fab after intravitreal injection in rabbits were taken from Gadkar et al. (2015). A least-squares method was used to estimate Dret and DRPE-cho with the 3D finite element model where mass transport was defined with diffusion and convection. Different intraocular pressures (IOP), initial distribution volumes (Vinit), and neural retina/vitreous partition coefficients (Kret/vit) were tested. Sensitivity analysis was performed for the final model. RESULTS With the final IgG model (IOP 10.1 Torr, Vinit 400 μl, Kret/vit 0.5), the estimated Dret and DRPE-cho were 36.8 × 10-9 cm2s-1 and 4.11 × 10-9 cm2s-1, respectively, and 76% of the dose was eliminated via the anterior chamber. Modeling of Fab revealed that a physiological model parameter "aqueous humor formation rate" sets constraints that need to be considered in the parameter estimation. CONCLUSIONS This study extends the use of 3D ocular PK models for parameter estimation using simultaneously macromolecule concentrations in three ocular tissues.
Collapse
Affiliation(s)
- Marko Lamminsalo
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland.
| | | | - Astrid Subrizi
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, St. Petersburg, Russian Federation
| | - Veli-Pekka Ranta
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| |
Collapse
|
41
|
Karachrysafi S, Sioga A, Komnenou A, Karamitsos A, Xioteli M, Dori I, Delis G, Kofidou E, Anastasiadou P, Sotiriou S, Karampatakis V, Papamitsou T. Histological Effects of Intravitreal Injection of Antifungal Agents in New Zealand White Rabbits: An Electron Microscopic and Immunohistochemical Study. Pharmaceuticals (Basel) 2020; 13:ph13100267. [PMID: 32977587 PMCID: PMC7598222 DOI: 10.3390/ph13100267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 11/16/2022] Open
Abstract
Fungal endophthalmitis is a serious and vision-threatening infection which requires an immediate and effective treatment approach. Our research aims to elucidate the histological effects of the intravitreal injection of the maximum safe dosage of voriconazole and micafungin on retina. Six albino New Zealand White Rabbits were used. In experimental animals, a solution of voriconazole (Group V) or micafungin (Group M) was intravitreally injected in the right eye, while in control animals, balanced salt solution was intravitreally injected in the left eye (Group C). Euthanasia was performed ten days post injection and the retina was removed and prepared for histological examination with a light and electron microscope. Eosin-hematoxylin staining did not reveal any pathological changes in any of the samples examined. The immunohistochemical staining for Tumor Necrosis Factor alpha (TNF-a) marker was detected as negative in all samples, while Interleukin 6 (IL-6) marker was detected as mild only in the group injected with voriconazole. Electron microscopy revealed several ultrastructural alterations in retinal layers in both groups of experimental animals. Histological retinal lesions, revealed with electron microscopy in the present investigation, raises the question of the safe usage of these antifungal agents in the treatment of fungal intraocular infections in the future.
Collapse
Affiliation(s)
- Sofia Karachrysafi
- Laboratory of Histology-Embryology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.S.); (T.P.)
- Correspondence:
| | - Antonia Sioga
- Laboratory of Histology-Embryology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.S.); (T.P.)
| | - Anastasia Komnenou
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.K.); (E.K.)
| | - Athanasios Karamitsos
- 2nd University Eye Clinic, Papageorgiou Hospital, Aristotle University of Thessaloniki, Greece: Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece;
| | - Maria Xioteli
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.X.); (I.D.)
| | - Ioanna Dori
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.X.); (I.D.)
| | - Georgios Delis
- Laboratory of Pharmacology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Evangelia Kofidou
- School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.K.); (E.K.)
| | - Penelope Anastasiadou
- Department of Oral Medicine/Pathology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Sotiris Sotiriou
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Vasileios Karampatakis
- Laboratory of Experimental Ophthalmology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Theodora Papamitsou
- Laboratory of Histology-Embryology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.S.); (T.P.)
| |
Collapse
|
42
|
Preclinical challenges for developing long acting intravitreal medicines. Eur J Pharm Biopharm 2020; 153:130-149. [DOI: 10.1016/j.ejpb.2020.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/01/2020] [Accepted: 05/08/2020] [Indexed: 02/07/2023]
|
43
|
Ilochonwu BC, Urtti A, Hennink WE, Vermonden T. Intravitreal hydrogels for sustained release of therapeutic proteins. J Control Release 2020; 326:419-441. [PMID: 32717302 DOI: 10.1016/j.jconrel.2020.07.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022]
Abstract
This review highlights how hydrogel formulations can improve intravitreal protein delivery to the posterior segment of the eye in order to increase therapeutic outcome and patient compliance. Several therapeutic proteins have shown excellent clinical successes for the treatment of various intraocular diseases. However, drug delivery to the posterior segment of the eye faces significant challenges due to multiple physiological barriers preventing drugs from reaching the retina, among which intravitreal protein instability and rapid clearance from the site of injection. Hence, frequent injections are required to maintain therapeutic levels. Moreover, because the world population ages, the number of patients suffering from ocular diseases, such as age-related macular degeneration (AMD) and diabetic retinopathy (DR) is increasing and causing increased health care costs. Therefore, there is a growing need for suitable delivery systems able to tackle the current limitations in retinal protein delivery, which also may reduce costs. Hydrogels have shown to be promising delivery systems capable of sustaining release of therapeutic proteins and thus extending their local presence. Here, an extensive overview of preclinically developed intravitreal hydrogels is provided with attention to the rational design of clinically useful intravitreal systems. The currently used polymers, crosslinking mechanisms, in vitro/in vivo models and advancements are discussed together with the limitations and future perspective of these biomaterials.
Collapse
Affiliation(s)
- Blessing C Ilochonwu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Arto Urtti
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
44
|
Kiss S, Grishanin R, Nguyen A, Rosario R, Greengard JS, Nieves J, Gelfman CM, Gasmi M. Analysis of Aflibercept Expression in NHPs following Intravitreal Administration of ADVM-022, a Potential Gene Therapy for nAMD. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:345-353. [PMID: 32671137 PMCID: PMC7341454 DOI: 10.1016/j.omtm.2020.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022]
Abstract
Several standard-of-care therapies for the treatment of retinal disease, including aflibercept, inhibit vascular endothelial growth factor (VEGFA). The main shortcoming of these therapies is potential undertreatment due to a lack of compliance resulting from the need for repeated injections. Gene therapy may provide sustained levels of anti-VEGFA proteins in the retina following a single injection. In this nonhuman primate study, we explored whether ADVM-022, a recombinant adeno-associated virus (AAV) vector designed to express aflibercept, could induce anti-VEGFA protein levels comparable with those observed following a single-bolus intravitreal (IVT) injection of the standard-of-care aflibercept recombinant protein. The results demonstrated that intraocular levels of aflibercept measured at 56 days after a single IVT injection of ADVM-022 were equivalent to those in the aflibercept recombinant protein-injected animals measured 21–32 days post-administration. ADVM-022-injected animals exhibited signs of an initial self-limiting inflammatory response, but overall all doses were well tolerated. ADVM-022 administration did not result in systemic exposure to aflibercept at any dose evaluated. These results demonstrated that a single IVT injection of ADVM-022 resulted in safe and efficacious aflibercept levels in the therapeutic range, suggesting the potential of a gene therapy approach for long-term treatment of retinal disease with anti-VEGF therapy.
Collapse
Affiliation(s)
- Szilárd Kiss
- Weill Cornell Medical College, New York, NY, USA
| | | | | | | | | | | | | | - Mehdi Gasmi
- Adverum Biotechnologies, Menlo Park, CA, USA
| |
Collapse
|
45
|
Smith DW, Lee CJ, Gardiner BS. No flow through the vitreous humor: How strong is the evidence? Prog Retin Eye Res 2020; 78:100845. [PMID: 32035123 DOI: 10.1016/j.preteyeres.2020.100845] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 02/06/2023]
Abstract
When analyzing vitreal drug delivery, or the pharmacological effects of drugs on intraocular pressure, or when interpreting outflow facility measurements, it is generally accepted that the fluid in the vitreous humor is stagnant. It is accepted that for all practical purposes, the aqueous fluid exits the eye via anterior pathways only, and so there is negligible if any posteriorly directed flow of aqueous through the vitreous humor. This assumption is largely based on the interpretation of experimental data from key sources including Maurice (1957), Moseley (1984), Gaul and Brubaker (1986), Maurice (1987) and Araie et al. (1991). However, there is strong independent evidence suggesting there is a substantial fluid flow across the retinal pigment epithelium from key sources including Cantrill and Pederson (1984), Chihara and Nao-i, Tsuboi (1985), Dahrouj et al. (2014), Smith and Gardiner (2017) and Smith et al. (2019). The conflicting evidence creates a conundrum-how can both interpretations be true? This leads us to re-evaluate the evidence. We demonstrate that the data believed to be supporting no aqueous flow through the vitreous are in fact compatible with a significant normal aqueous flow. We identify strong and independent lines of evidence supporting fluid flow across the RPE, including our new outflow model for the eye. On balance it appears the current evidence favors the view that there is normally a significant aqueous flow across the RPE in vivo. This finding suggests that past and future analyses of outflow facility, interpretations of some drug distributions and the interpretation of some drug effects on eye tissues, may need to be revised.
Collapse
Affiliation(s)
- David W Smith
- Faculty of Engineering and Mathematical Sciences, The University of Western Australia, Perth, Australia.
| | - Chang-Joon Lee
- Faculty of Engineering and Mathematical Sciences, The University of Western Australia, Perth, Australia; College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Western Australia, Australia
| | - Bruce S Gardiner
- College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Western Australia, Australia
| |
Collapse
|
46
|
Caruso A, Füth M, Alvarez-Sánchez R, Belli S, Diack C, Maass KF, Schwab D, Kettenberger H, Mazer NA. Ocular Half-Life of Intravitreal Biologics in Humans and Other Species: Meta-Analysis and Model-Based Prediction. Mol Pharm 2020; 17:695-709. [PMID: 31876425 DOI: 10.1021/acs.molpharmaceut.9b01191] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutic antibodies administered intravitreally are the current standard of care to treat retinal diseases. The ocular half-life (t1/2) is a key determinant of the duration of target suppression. To support the development of novel, longer-acting drugs, a reliable determination of t1/2 is needed together with an improved understanding of the factors that influence it. A model-based meta-analysis was conducted in humans and nonclinical species (rat, rabbit, monkey, and pig) to determine consensus values for the ocular t1/2 of IgG antibodies and Fab fragments. Results from multiple literature and in-house pharmacokinetic studies are presented within a mechanistic framework that assumes diffusion-controlled drug elimination from the vitreous. Our analysis shows, both theoretically and experimentally, that the ocular t1/2 increases in direct proportion to the product of the hydrodynamic radius of the macromolecule (3.0 nm for Fab and 5.0 nm for IgG) and the square of the radius of the vitreous globe, which varies approximately 24-fold from the rat to the human. Interspecies differences in the proportionality factors are observed and discussed in mechanistic terms. In addition, mathematical formulae are presented that allow prediction of the ocular t1/2 for molecules of interest. The utility of these formulae is successfully demonstrated in case studies of aflibercept, brolucizumab, and PEGylated Fabs, where the predicted ocular t1/2 values are found to be in reasonable agreement with the experimental data available for these molecules.
Collapse
Affiliation(s)
- Antonello Caruso
- Pharmaceutical Sciences, Roche Innovation Center Basel , Roche Pharma Research and Early Development , Basel 4070 , Switzerland
| | - Matthias Füth
- Pharmaceutical Sciences, Roche Innovation Center Basel , Roche Pharma Research and Early Development , Basel 4070 , Switzerland
| | - Ruben Alvarez-Sánchez
- Pharmaceutical Sciences, Roche Innovation Center Basel , Roche Pharma Research and Early Development , Basel 4070 , Switzerland
| | - Sara Belli
- Pharmaceutical Sciences, Roche Innovation Center Basel , Roche Pharma Research and Early Development , Basel 4070 , Switzerland
| | - Cheikh Diack
- Pharmaceutical Sciences, Roche Innovation Center Basel , Roche Pharma Research and Early Development , Basel 4070 , Switzerland
| | - Katie F Maass
- Clinical Pharmacology , Genentech , South San Francisco 94080 , California , United States
| | - Dietmar Schwab
- Pharmaceutical Sciences, Roche Innovation Center Basel , Roche Pharma Research and Early Development , Basel 4070 , Switzerland
| | - Hubert Kettenberger
- Therapeutic Modalities, Roche Innovation Center Munich , Roche Pharma Research and Early Development , Penzberg 82377 , Germany
| | - Norman A Mazer
- Pharmaceutical Sciences, Roche Innovation Center Basel , Roche Pharma Research and Early Development , Basel 4070 , Switzerland
| |
Collapse
|
47
|
Penkova AN, Zhang S, Humayun MS, Fraser S, Moats R, Singh Sadhal S. MEASUREMENT OF THE HYDRAULIC CONDUCTIVITY OF THE VITREOUS HUMOR. JOURNAL OF POROUS MEDIA 2020; 23:195-206. [PMID: 32494116 PMCID: PMC7269170 DOI: 10.1615/jpormedia.2020028229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The hydraulic conductivity of the vitreous humor has been measured for the bovine eye. The experiment was carried out by placing it within upright cylindrical chamber, open at both ends, and letting its liquid content drain out of the bottom opening by gravity, through a 20μm nylon mesh filter. Additional negative pressure was provided at the exit by a hanging drainage tube. The diminishing vitreous volume was measured in terms of the height in the chamber and recorded as a function of time. The reduction in the vitreous liquid content also caused the hydraulic conductivity to reduce and this parameter was quantified on the basis of previously-developed theories of fibrous porous media that have been very well established. A theoretical model with a fully analytical expression for the vitreous volume undergoing drainage was developed and used as a least-squares best fit to deliver the initial hydraulic conductivity value of K 0/μ=(7.8 ± 3.1) × 10-12 m2 (Pa-s). The measurements were made with the hyaloid membrane intact and therefore represents an effective conductivity for the entire system, including possible variations within the vitreous.
Collapse
Affiliation(s)
- Anita N Penkova
- Department of Aerospace & Mechanical Engineering, University of Southern California, USC Viterbi School of Engineering, Los Angeles, CA 90089-1453
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027
| | - Shuqi Zhang
- Department of Aerospace & Mechanical Engineering, University of Southern California, USC Viterbi School of Engineering, Los Angeles, CA 90089-1453
| | - Mark S Humayun
- Department of Ophthalmology, USC Roski Eye Institute, Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033-4682
- Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089-1111
| | - Scott Fraser
- Departments of Molecular and Computational Biology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90089-0371
- Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089-1111
| | - Rex Moats
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027
- Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089-1111
| | - Satwindar Singh Sadhal
- Department of Aerospace & Mechanical Engineering, University of Southern California, USC Viterbi School of Engineering, Los Angeles, CA 90089-1453
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033-4682
| |
Collapse
|
48
|
Crowell SR, Wang K, Famili A, Shatz W, Loyet KM, Chang V, Liu Y, Prabhu S, Kamath AV, Kelley RF. Influence of Charge, Hydrophobicity, and Size on Vitreous Pharmacokinetics of Large Molecules. Transl Vis Sci Technol 2019; 8:1. [PMID: 31695962 PMCID: PMC6827426 DOI: 10.1167/tvst.8.6.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/24/2019] [Indexed: 01/04/2023] Open
Abstract
Purpose Development of therapeutics for retinal disease with improved durability is hampered by inadequate understanding of pharmacokinetic (PK) drivers following intravitreal injection. Previous work shows that hydrodynamic radius is correlated with vitreal half-life over the range of 3 to 7 nm, and that charge and hydrophobicity influence systemic clearance. Better understanding the molecular attributes affecting vitreal elimination half-life enables improved design of therapeutics and enhances clinical translatability. Methods Impacts of charge and hydrophobicity on vitreal PK in the rabbit were systematically assessed using antibody and antibody fragment (Fab) variant series, including ranibizumab, altered through amino acid changes in hypervariable regions of the light chain. The impact of molecule size on vitreal PK was assessed in the rabbit, nonhuman primate, and human for a range of molecules (1–45 nm, net charge −1324 to +22.9 in rabbit), including published and internal data. Results No correlation was observed between vitreal PK and charge or hydrophobicity. Equivalent rabbit vitreal PK was observed for ranibizumab and its variants with isoelectric points (pI) in the range of 6.8 to 10.2, and hydrophobicities of the variable domain unit (FvHI) between 1009 and 1296; additional variant series had vitreal PK similarly unaffected by pI (5.4–10.2) and FvHI (1004–1358). Strong correlations were observed between vitreal half-life and hydrodynamic radius for preclinical species (R2 = 0.8794–0.9366). Conclusions Diffusive properties of soluble large molecules, as quantified by hydrodynamic radius, make a key contribution to vitreal elimination, whereas differences in charge or hydrophobicity make minor or negligible contributions. Translational Relevance These results support estimation of vitreal elimination rates based on molecular size in relevant preclinical species and humans.
Collapse
Affiliation(s)
- Susan R Crowell
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, CA, USA
| | - Kathryn Wang
- Drug Delivery, Genentech, South San Francisco, CA, USA
| | - Amin Famili
- Drug Delivery, Genentech, South San Francisco, CA, USA
| | - Whitney Shatz
- Protein Chemistry, Genentech, South San Francisco, CA, USA
| | - Kelly M Loyet
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Vincent Chang
- Bioanalytical Sciences, Genentech, South San Francisco, CA, USA
| | - Yanqiu Liu
- Bioanalytical Sciences, Genentech, South San Francisco, CA, USA
| | - Saileta Prabhu
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, CA, USA
| | - Amrita V Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, CA, USA
| | | |
Collapse
|
49
|
Investigating impacts of surface charge on intraocular distribution of intravitreal lipid nanoparticles. Exp Eye Res 2019; 186:107711. [DOI: 10.1016/j.exer.2019.107711] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/12/2019] [Accepted: 06/21/2019] [Indexed: 12/15/2022]
|
50
|
Huang X, Chau Y. Intravitreal nanoparticles for retinal delivery. Drug Discov Today 2019; 24:1510-1523. [DOI: 10.1016/j.drudis.2019.05.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/17/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
|