1
|
Sorsa T, Del Amo EM, Sadeghi A, Laitinen J, Pilipenko I, Urtti A, Subrizi A. Novel in vitro model for intravitreal drug dissolution and release studies of small molecules and their long-acting formulations. J Control Release 2025; 383:113823. [PMID: 40339659 DOI: 10.1016/j.jconrel.2025.113823] [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: 03/03/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Intravitreal drug delivery is essential for multiple ocular diseases. Development of intravitreal formulations requires high-quality in vivo experiments that require a large number of animals. This development can be expedited by improving in vitro testing and utilizing in vitro - in vivo correlation. We propose an in vitro approach using vitreous in a dialysis membrane to conduct these studies. The model was developed for small molecules and evaluated with drugs in solution (13 drugs in one cassette and 4 in another), and a drug suspension. For solutions, 6 out of 8 drug compounds were within 2.5-fold difference to their experimental vitreal clearances. Furthermore, we demonstrated suitability of the model for long-acting formulations in a 150-day experiment with triamcinolone acetonide suspension. The suspension displayed similar elimination trend as has been observed in rabbits. We believe this system can lead the way in introducing site-specific drug release testing for the intravitreal route.
Collapse
Affiliation(s)
- Teemu Sorsa
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3 C, 70210 Kuopio, Finland
| | - Eva M Del Amo
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3 C, 70210 Kuopio, Finland
| | - Amir Sadeghi
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3 C, 70210 Kuopio, Finland
| | - Jonna Laitinen
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3 C, 70210 Kuopio, Finland
| | - Iuliia Pilipenko
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3 C, 70210 Kuopio, Finland
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3 C, 70210 Kuopio, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, University of Helsinki, 00014 Helsinki, Finland
| | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, Yliopistonrinne 3 C, 70210 Kuopio, Finland
| |
Collapse
|
2
|
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. J Pharm Sci 2025; 114:1164-1174. [PMID: 39694270 PMCID: PMC11932321 DOI: 10.1016/j.xphs.2024.12.005] [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: 08/18/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
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 in the vitreous. The rabbit and human models predict bevacizumab concentration in the vitreous and fovea, enhancing knowledge and understanding of wet AMD treatment.
Collapse
Affiliation(s)
- Jabia M Chowdhury
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Department of Electrical Engineering, Texas A&M University-Texarkana, Texarkana, TX, 75503, USA
| | - Eduardo A Chacin Ruiz
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Matthew P Ohr
- Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, 43212, USA
| | - Katelyn E Swindle-Reilly
- Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, 43212, USA; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA; William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Ashlee N Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA; Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14215, USA.
| |
Collapse
|
3
|
Khan MS, Murthy A, Ahmed T. Advancements in Ocular Modelling and Simulations: Key Considerations and Case Studies. AAPS PharmSciTech 2024; 26:14. [PMID: 39690355 DOI: 10.1208/s12249-024-03001-7] [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: 08/14/2024] [Accepted: 11/12/2024] [Indexed: 12/19/2024] Open
Abstract
This review paper discusses the key aspects of ocular biopharmaceutics, with emphasis on the crucial role played by ocular compartmental modelling and simulation in deciphering physiological conditions related to various eye diseases. It describes eye's intricate structure and function and the need for precise and targeted drug delivery systems to address prevalent eye conditions. The review categorizes and discusses various formulations employed in ocular drug delivery, delineating their respective advantages and limitations. Additionally, it probes the challenges inherent in diverse routes of drug administration for ocular therapies and provides insights into the complexities of achieving optimal drug concentrations at the target site within the eye. The central theme of this work is the ocular compartmental modelling and simulations. Hence, this works discusses on the nuanced understanding of physiological conditions within the eye, drug distribution, drug release kinetics, and key considerations for ocular compartmental modelling and simulations. By combining information from various sources, this review aims to serve as a comprehensive reference for researchers, clinicians, and pharmaceutical developers. It covers the multifaceted landscape of ocular biopharmaceutics and the transformative impact of modelling and simulation in optimizing ocular drug delivery strategies.
Collapse
Affiliation(s)
- Mohammed Shareef Khan
- Biopharmaceutics - Biopharmaceutics and Bioequivalence, Global Clinical Management, Dr. Reddy's Laboratories Ltd, Hyderabad, India.
| | - Aditya Murthy
- Biopharmaceutics - Biopharmaceutics and Bioequivalence, Global Clinical Management, Dr. Reddy's Laboratories Ltd, Hyderabad, India
| | - Tausif Ahmed
- Biopharmaceutics and Bioanalytical - Global Clinical Management, Dr. Reddy's Laboratories Ltd, Hyderabad, India
| |
Collapse
|
4
|
Chowdhury JM, Ruiz EAC, 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 in the vitreous. The rabbit and human models predict bevacizumab concentration in the vitreous and fovea, enhancing knowledge and understanding of wet AMD treatment.
Collapse
|
5
|
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
|
6
|
Naware S, Bussing D, Shah DK. Translational physiologically-based pharmacokinetic model for ocular disposition of monoclonal antibodies. J Pharmacokinet Pharmacodyn 2024; 51:493-508. [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] [MESH Headings] [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
|
7
|
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
|
8
|
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: 1.0] [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
|
9
|
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: 7] [Impact Index Per Article: 3.5] [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
|
10
|
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: 6] [Impact Index Per Article: 3.0] [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
|
11
|
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: 7] [Impact Index Per Article: 2.3] [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
|
12
|
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: 0.3] [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
|
13
|
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]
|
14
|
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
|
15
|
Huerta Ángeles G, Nešporová K. Hyaluronan and its derivatives for ophthalmology: Recent advances and future perspectives. Carbohydr Polym 2021; 259:117697. [PMID: 33673986 DOI: 10.1016/j.carbpol.2021.117697] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/26/2022]
|
16
|
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: 64] [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
|
17
|
Abstract
Clear vision is dependent on features that protect the anatomical integrity of the eye (cornea and sclera) and those that contribute to internal ocular homeostasis by conferring hemangiogenic (avascular tissues and antiangiogenic factors), lymphangiogenic (lack of draining lymphatics), and immunologic (tight junctions that form blood-ocular barriers, immunosuppressive cells, and modulators) privileges. The later examples are necessary components that enable the eye to maintain an immunosuppressive environment that responds to foreign invaders in a deviated manner, minimizing destructive inflammation that would impair vision. These conditions allowed for the observations made by Medawar, in 1948, of delayed rejection of allogenic tissue grafts in the anterior chamber of mouse eye and permit the sequestration of foreign invaders (eg, Toxoplasma gondii) within the retina of healthy individuals. Yet successful development of intraocular drugs (biologics and delivery devices) has been stymied by adverse ocular pathology, much of which is driven by immune pathways. The eye can be intolerant of foreign protein irrespective of delivery route, and endogenous ocular cells have remarkable plasticity when recruited to preserve visual function. This article provides a review of current understanding of ocular immunology and the potential role of immune mechanisms in pathology observed with intraocular drug delivery.
Collapse
Affiliation(s)
| | | | - Sharmila Masli
- 12259Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
18
|
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: 0.4] [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
|
19
|
Le Merdy M, Spires J, Lukacova V, Tan ML, Babiskin A, Xu X, Zhao L, Bolger MB. Ocular Physiologically Based Pharmacokinetic Modeling for Ointment Formulations. Pharm Res 2020; 37:245. [PMID: 33215336 PMCID: PMC7677276 DOI: 10.1007/s11095-020-02965-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/27/2020] [Indexed: 12/26/2022]
Abstract
Purpose The purpose of this study is to show how the Ocular Compartmental Absorption & Transit (OCAT™) model in GastroPlus® can be used to characterize ocular drug pharmacokinetic performance in rabbits for ointment formulations. Methods A newly OCAT™ model developed for fluorometholone, as well as a previously verified model for dexamethasone, were used to characterize the aqueous humor (AH) concentration following the administration of multiple ointment formulations to rabbit. The model uses the following parameters: application surface area (SA), a fitted application time, and the fitted Higuchi release constant to characterize the rate of passage of the active pharmaceutical ingredient from the ointment formulations into the tears in vivo. Results Parameter sensitivity analysis was performed to understand the impact of ointment formulation changes on ocular exposure. While application time was found to have a significant impact on the time of maximal concentration in AH, both the application SA and the Higuchi release constant significantly influenced both the maximum concentration and the ocular exposure. Conclusions This initial model for ointment ophthalmic formulations is a first step to better understand the interplay between physiological factors and ophthalmic formulation physicochemical properties and their impact on in vivo ocular drug pharmacokinetic performance in rabbits. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-020-02965-y.
Collapse
Affiliation(s)
- Maxime Le Merdy
- Simulations Plus, Inc., 42505 10th Street West, Lancaster, California, 93534, USA.
| | - Jessica Spires
- Simulations Plus, Inc., 42505 10th Street West, Lancaster, California, 93534, USA
| | - Viera Lukacova
- Simulations Plus, Inc., 42505 10th Street West, Lancaster, California, 93534, USA
| | - Ming-Liang Tan
- Food and Drug Administration, CDER/OGD/ORS/DQMM, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Andrew Babiskin
- Food and Drug Administration, CDER/OGD/ORS/DQMM, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Xiaoming Xu
- Food and Drug Administration, CDER/OPQ/OTR/DPQR, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Liang Zhao
- Food and Drug Administration, CDER/OGD/ORS/DQMM, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Michael B Bolger
- Simulations Plus, Inc., 42505 10th Street West, Lancaster, California, 93534, USA
| |
Collapse
|
20
|
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: 0.6] [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
|
21
|
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: 87] [Impact Index Per Article: 17.4] [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
|
22
|
Oh WT, Lee JB, Choi W, Bae HW, Kim CS, Kim CY, Sung HJ. Shape Memory Tube Plug for Fine-control of Intraocular Pressure by Glaucoma Devices. ACS Biomater Sci Eng 2020; 6:3784-3790. [PMID: 33463360 DOI: 10.1021/acsbiomaterials.0c00649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
As a standard approach to treating glaucoma, a silicone tube device is inserted into the eye to drain water and thereby lower the intraocular pressure (IOP). However, the fixed diameter of the tube often results in too much initial water drain. Suture-sheaving of the outer tube wall also leads to random IOP drops over time. In this study, self-expansion of the inner tube diameter was achieved by inserting a shape memory tube into the silicone tube. The difference in controlling small to large IOP drops before and after tube diameter expansion was demonstrated via computational modeling, a flow pumping system, and rabbit experiments.
Collapse
Affiliation(s)
- Won Taek Oh
- TMD LAB Co., Ltd., Seoul 03722, Republic of Korea
| | - Jung Bok Lee
- Department of Biological Science, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul 04310, Republic of Korea
| | - Wungrak Choi
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyoung Won Bae
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | | | - Chan Yun Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Hak-Joon Sung
- Medical Engineering, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Republic of Korea
| |
Collapse
|
23
|
Heikkinen EM, Ruponen M, Jasper LM, Leppänen J, Hellinen L, Urtti A, Auriola S, Rautio J, Vellonen KS. Prodrug Approach for Posterior Eye Drug Delivery: Synthesis of Novel Ganciclovir Prodrugs and in Vitro Screening with Cassette Dosing. Mol Pharm 2020; 17:1945-1953. [PMID: 32320251 DOI: 10.1021/acs.molpharmaceut.0c00037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Because of poor ocular drug bioavailability, intravitreal injections have become the gold standard for drug delivery to the posterior eye. The prodrug approach can be used for optimizing the biopharmaceutical properties of intravitreal drugs. The preclinical screening of prodrugs' properties, such as hydrolysis and bioconversion, should be conducted in a resource-efficient way for an extensive set of synthesized compounds with validated methods. Our objective was to explore cassette dosing in in vitro prodrug hydrolysis and bioconversion studies in buffer, vitreous, and retinal pigment epithelium (RPE) homogenate for rapid medium-throughput screening. Moreover, our aim was to correlate the prodrug structure with hydrolytic behavior. We synthesized 18 novel ganciclovir prodrugs and first studied their hydrolysis in aqueous buffer and porcine vitreous in vitro with cassette dosing for 35 h. A method for vitreous homogenate pH equilibration to a physiological level by using buffer and incubation under 5% carbon dioxide was validated. The hydrolysis of the prodrugs was evaluated in porcine RPE homogenate in vitro with cassette dosing, and five prodrugs were assayed individually to examine their bioconversion into ganciclovir in RPE after 2 h. Lastly, the prodrugs' binding to melanin was studied in vitro. The prodrugs showed a wide spectrum of hydrolysis rates, ranging from a few percentages to 100% in the vitreous and RPE; in general, hydrolysis in RPE was faster than in vitreous. Prodrugs with long carbon chains and disubstitution showed lability in the tissue homogenates, whereas prodrugs with branched carbon chains and aromatic groups were stable. All five prodrugs chosen for the bioconversion study in RPE were hydrolyzed into ganciclovir, and their hydrolytic behavior matched results from the cassette mix experiment, supporting the cassette mix approach for hydrolysis and bioconversion studies. None of the prodrugs bound highly to melanin (<50% bound). In conclusion, cassette dosing proved useful for the rapid screening of prodrug hydrolysis and bioconversion properties. Analyzing several compounds simultaneously can complicate the analytics, and thus, choosing the compounds of the cassette mix should be done carefully to avoid mutual interference of the compounds with the results. The methodology and results of the work are applicable in ocular drug research and prodrug design.
Collapse
Affiliation(s)
- Emma M Heikkinen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| | - Lisa-Marie Jasper
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| | - Jukka Leppänen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| | - Laura Hellinen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland.,Helsingin Yliopisto, Faculty of Pharmacy, Division of Pharmaceutical Biosciences, Viikinkaari 5E, Helsinki 00014, Finland.,Institute of Chemistry, Saint Petersburg State University, Universitetskii Prospect 26, Sankt-Peterburg 198504, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| | - Jarkko Rautio
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| | - Kati-Sisko Vellonen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, Kuopio 70211, Finland
| |
Collapse
|
24
|
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: 6.2] [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
|
25
|
He L, Wang H, Han Y, Wang K, Dong H, Li Y, Shi D, Li Y. Remodeling of Cellular Surfaces via Fast Disulfide-Thiol Exchange To Regulate Cell Behaviors. ACS APPLIED MATERIALS & INTERFACES 2019; 11:47750-47761. [PMID: 31773939 DOI: 10.1021/acsami.9b17550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Remodeling of cellular surfaces is shown highly effective in the manipulation and control of cell behaviors via nonbiological means. By 5-thio-2-nitrobenzoate-mediated, fast, and reversible disulfide-thiol exchange, a sequential layer by layer assembly process was developed to grow albumin protein shells on cellular surfaces fixed by a disulfide-linked network, in a cytocompatible manner. The artificial shells, accomplished by a double-assembly process, were sustainable up to >1 day, and thereafter gradually bioabsorbed with unaffected cell viability. The surface engineering process enabled dynamic remodeling of cellular surfaces that effectively controlled cell behaviors including regulated cell proliferation, enhanced uptake efficiency of dextran-fluorescein isothiocyanate that is known for cell-impermeability, and targeted imaging. This unique approach was well-validated on tumor cells (B16), immune cells (DC2.4), and neutrophils, showing its potential universality for most of the cells that are rich in thiols. The new strategy will show promise in cell manipulation and targeted imaging.
Collapse
Affiliation(s)
- Lianghua He
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Huaiji Wang
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Yi Han
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Kun Wang
- School of Materials Science and Engineering , Tongji University , 4800 Caoan Road , Shanghai 201804 , China
| | - Haiqing Dong
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Yan Li
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| | - Donglu Shi
- The Materials Science & Engineering Program, Department of Mechanical & Materials Engineering, College of Engineering & Applied Science , University of Cincinnati , Cincinnati , Ohio 45221 , United States
| | - Yongyong Li
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , China
| |
Collapse
|
26
|
Schmitt M, Hippeläinen E, Raviña M, Arango-Gonzalez B, Antopolsky M, Vellonen KS, Airaksinen AJ, Urtti A. Intravitreal Pharmacokinetics in Mice: SPECT/CT Imaging and Scaling to Rabbits and Humans. Mol Pharm 2019; 16:4399-4404. [DOI: 10.1021/acs.molpharmaceut.9b00679] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Mechthild Schmitt
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Eero Hippeläinen
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- HUS Medical imaging Center, Clinical Physiology and Nuclear Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Manuela Raviña
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Maxim Antopolsky
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Anu J. Airaksinen
- Department of Chemistry—Radiochemistry, University of Helsinki, Helsinki, Finland
| | - Arto Urtti
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, Peterhoff, Russian Federation
| |
Collapse
|
27
|
Henein C, Awwad S, Ibeanu N, Vlatakis S, Brocchini S, Tee Khaw P, Bouremel Y. Hydrodynamics of Intravitreal Injections into Liquid Vitreous Substitutes. Pharmaceutics 2019; 11:E371. [PMID: 31374925 PMCID: PMC6723562 DOI: 10.3390/pharmaceutics11080371] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 01/08/2023] Open
Abstract
Intravitreal injections have become the cornerstone of retinal care and one of the most commonly performed procedures across all medical specialties. The impact of hydrodynamic forces of intravitreal solutions when injected into vitreous or vitreous substitutes has not been well described. While computational models do exist, they tend to underestimate the starting surface area of an injected bolus of a drug. Here, we report the dispersion profile of a dye bolus (50 µL) injected into different vitreous substitutes of varying viscosities, surface tensions, and volumetric densities. A novel 3D printed in vitro model of the vitreous cavity of the eye was designed to visualize the dispersion profile of solutions when injected into the following vitreous substitutes-balanced salt solution (BSS), sodium hyaluronate (HA), and silicone oils (SO)-using a 30G needle with a Reynolds number (Re) for injection ranging from approximately 189 to 677. Larger bolus surface areas were associated with faster injection speeds, lower viscosity of vitreous substitutes, and smaller difference in interfacial surface tensions. Boluses exhibited buoyancy when injected into standard S1000. The hydrodynamic properties of liquid vitreous substitutes influence the initial injected bolus dispersion profile and should be taken into account when simulating drug dispersion following intravitreal injection at a preclinical stage of development, to better inform formulations and performance.
Collapse
Affiliation(s)
- Christin Henein
- Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust and UCL Institute of Ophthalmology, National Institute for Health Research (NIHR), London EC1V 9EL, UK
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Sahar Awwad
- Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust and UCL Institute of Ophthalmology, National Institute for Health Research (NIHR), London EC1V 9EL, UK
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Nkiruka Ibeanu
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Stavros Vlatakis
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Steve Brocchini
- Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust and UCL Institute of Ophthalmology, National Institute for Health Research (NIHR), London EC1V 9EL, UK
- School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Peng Tee Khaw
- Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust and UCL Institute of Ophthalmology, National Institute for Health Research (NIHR), London EC1V 9EL, UK
| | - Yann Bouremel
- Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust and UCL Institute of Ophthalmology, National Institute for Health Research (NIHR), London EC1V 9EL, UK.
- School of Pharmacy, University College London, London WC1N 1AX, UK.
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK.
| |
Collapse
|
28
|
Missel PJ, Sarangapani R. Physiologically based ocular pharmacokinetic modeling using computational methods. Drug Discov Today 2019; 24:1551-1563. [PMID: 31319151 DOI: 10.1016/j.drudis.2019.05.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/08/2019] [Accepted: 05/31/2019] [Indexed: 01/21/2023]
Abstract
By explicitly representing ocular anatomy, computational fluid dynamic simulation methods model drug mass transport both within and between ocular tissue regions, providing reliable animal-to-human translation of bioavailability. Here, we apply physiologically based models to simulate ocular drug administration. A non-anatomical model is used that applies a simple theorem for calculating ocular bioavailability from a topical dose. A computational fluid dynamic model is also described that incorporates ocular physiology in anatomical models for rabbit, monkey and man. This second method applies material properties and boundary conditions for various tissues enabling simulation of fluid flows, pressures, temperatures, convection, and drug advection following various modes of administration. The method provides a regional distribution with a given tissue not available using standard compartmental models, and enables translation of results from animal experiments into predictions for human ocular pharmacokinetics (PK).
Collapse
Affiliation(s)
- Paul J Missel
- Data Science and Digital Solutions, Alcon Vision LLC, Fort Worth, TX, USA.
| | - Ramesh Sarangapani
- Data Science and Digital Solutions, Alcon Vision LLC, Fort Worth, TX, USA
| |
Collapse
|
29
|
Le Merdy M, Fan J, Bolger MB, Lukacova V, Spires J, Tsakalozou E, Patel V, Xu L, Stewart S, Chockalingam A, Narayanasamy S, Rouse R, Matta M, Babiskin A, Kozak D, Choi S, Zhang L, Lionberger R, Zhao L. Application of Mechanistic Ocular Absorption Modeling and Simulation to Understand the Impact of Formulation Properties on Ophthalmic Bioavailability in Rabbits: a Case Study Using Dexamethasone Suspension. AAPS JOURNAL 2019; 21:65. [PMID: 31111305 DOI: 10.1208/s12248-019-0334-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Developing mathematical models to predict changes in ocular bioavailability and pharmacokinetics due to differences in the physicochemical properties of complex topical ophthalmic suspension formulations is important in drug product development and regulatory assessment. Herein, we used published FDA clinical pharmacology review data, in-house, and literature rabbit pharmacokinetic data generated for dexamethasone ophthalmic suspensions to demonstrate how the mechanistic Ocular Compartmental Absorption and Transit model by GastroPlus™ can be used to characterize ocular drug pharmacokinetic performance in rabbits for suspension formulations. This model was used to describe the dose-dependent (0.01 to 0.1%) non-linear pharmacokinetic in ocular tissues and characterize the impact of viscosity (1.67 to 72.9 cP) and particle size (5.5 to 22 μm) on in vivo ocular drug absorption and disposition. Parameter sensitivity analysis (hypothetical suspension particle size: 1 to 10 μm, viscosity: 1 to 100 cP) demonstrated that the interplay between formulation properties and physiological clearance through drainage and tear turnover rates in the pre-corneal compartment drives the ocular drug bioavailability. The quick removal of drug suspended particles from the pre-corneal compartment renders the impact of particle size inconsequential relative to viscosity modification. The in vivo ocular absorption is (1) viscosity non-sensitive when the viscosity is high and the impact of viscosity on the pre-corneal residence time reaches the maximum physiological system capacity or (2) viscosity sensitive when the viscosity is below a certain limit. This study reinforces our understanding of the interplay between physiological factors and ophthalmic formulation physicochemical properties and their impact on in vivo ocular drug PK performance in rabbits.
Collapse
Affiliation(s)
- Maxime Le Merdy
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Jianghong Fan
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA.
| | - Michael B Bolger
- Simulations Plus, Inc., 42505 10th Street West, Lancaster, California, 93534, USA
| | - Viera Lukacova
- Simulations Plus, Inc., 42505 10th Street West, Lancaster, California, 93534, USA
| | - Jessica Spires
- Simulations Plus, Inc., 42505 10th Street West, Lancaster, California, 93534, USA
| | - Eleftheria Tsakalozou
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Vikram Patel
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Lin Xu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Sharron Stewart
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Ashok Chockalingam
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Suresh Narayanasamy
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Rodney Rouse
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Murali Matta
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 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, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Darby Kozak
- Division of Therapeutic Performance, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Stephanie Choi
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| | - Robert Lionberger
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 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, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, Maryland, 20993, USA
| |
Collapse
|
30
|
Rimpelä AK, Kiiski I, Deng F, Kidron H, Urtti A. Pharmacokinetic Simulations of Intravitreal Biologicals: Aspects of Drug Delivery to the Posterior and Anterior Segments. Pharmaceutics 2018; 11:pharmaceutics11010009. [PMID: 30598037 PMCID: PMC6359489 DOI: 10.3390/pharmaceutics11010009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 12/21/2018] [Accepted: 12/24/2018] [Indexed: 12/15/2022] Open
Abstract
Biologicals are important ocular drugs that are be delivered using monthly and bimonthly intravitreal injections to treat retinal diseases, such as age-related macular degeneration. Long acting delivery systems are needed for prolongation of their dosing interval. Intravitreal biologicals are eliminated from the eye via the aqueous humor outflow. Thus, the anterior and posterior segments are exposed to the drug. We utilized a kinetic simulation model to estimate protein drug concentrations in the vitreous and aqueous humor after bolus injection and controlled release administration to the vitreous. The simulations predicted accurately the experimental levels of 5 biologicals in the vitreous and aqueous humor. The good match between the simulations and experimental data demonstrated almost complete anterior segment bioavailability, and major dose sparing with ocular controlled release systems. Overall, the model is a useful tool in the design of intraocular delivery of biologicals.
Collapse
Affiliation(s)
- Anna-Kaisa Rimpelä
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland.
| | - Iiro Kiiski
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland.
| | - Feng Deng
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland.
| | - Heidi Kidron
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland.
| | - Arto Urtti
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland.
- Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, Universitetskii pr. 26, Peterhoff, 198504 St. Petersburg, Russia.
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70211 Kuopio, Finland.
| |
Collapse
|