1
|
Ibeas Moreno E, Alonso MJ, Abbadessa A. Intraocular injectable hydrogels for the delivery of cells and nanoparticles. Mater Today Bio 2025; 32:101767. [PMID: 40290894 PMCID: PMC12033996 DOI: 10.1016/j.mtbio.2025.101767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
The rising global life expectancy has led to a growing prevalence of ophthalmic diseases, while current treatments face important limitations in terms of efficacy, costs, and patient compliance. The use of injectable hydrogels as drug and cell carriers is a promising approach, compared to the injection of drug solutions or cell suspensions. This is because the hydrogel matrix may offer protection against clearance or degradation, may modulate drug/cell release, and provide a biomimetic substrate for differentiating cells while being minimally invasive. On one hand, injectable hydrogels for ocular drug delivery have been traditionally designed to host and release small drugs or proteins. However, limitations such as high burst release and difficulty of entrapping hydrophobic molecules led to the emergence of nanocomposite hydrogels, where the drug is entrapped in nanoparticles prior hydrogel incorporation. Composite systems offer great advantages over the injection of particle suspensions, improving particle fate and drug release kinetics. On the other hand, injectable hydrogels offer a cell-friendly environment to seek tissue regeneration, providing biomechanical and biochemical cues for cellular cross-talk, differentiation, and formation of new extracellular matrix. This review critically discusses recent advancements in the development of novel injectable hydrogels as delivery vehicles for drug-loaded nanoparticles and cells, with a major focus on the formulation components, administration routes, and other factors affecting performance, highlighting promising aspects and challenges to address in the future.
Collapse
Affiliation(s)
- Elena Ibeas Moreno
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
- Madrid Institute for Advanced Studies in Nanoscience (IMDEA), 28049, Madrid, Spain
| | - Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Campus Vida, 15782, Santiago de Compostela, Spain
| |
Collapse
|
2
|
Minnaert AK, Nguyen VP, Harizaj A, Goemaere I, Van Puyvelde B, Dhaenens M, Sauvage F, Deforce D, Salmaso S, Caliceti P, Braeckmans K, Peynshaert K, Paulus YM, De Smedt SC, Remaut K. Collagenase-modified polydopamine nanoparticles for safe and effective vitreolysis. J Control Release 2025; 383:113753. [PMID: 40254139 DOI: 10.1016/j.jconrel.2025.113753] [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: 11/29/2024] [Revised: 03/29/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Anomalous posterior vitreous detachment (aPVD) is involved in the pathogenesis of many vitreoretinal disorders. This condition is currently managed by vitrectomy, a routine but invasive surgery which is especially challenging in patients with firm vitreoretinal adhesions. Since 1998, pharmacological vitreolysis emerged as a potential replacement or adjunct therapy to vitrectomy. Over the years, most attention was focused on enzymes, but their use has been limited mainly due to retinal toxicity. To revive the potential of enzymatic vitreolysis, we aimed to immobilize collagenase on the surface of polydopamine nanoparticles to prevent penetration into the retinal layers. We synthesized stable and functional collagenase-modified nanoparticles and were able to induce vitreous liquefaction and complete PVD ex vivo. Moreover, we demonstrated a substantial reduction in retinal toxicity upon injection in bovine vitreoretinal explants. Subsequent in vivo analysis revealed that retinal morphology and function were preserved, in contrast to free collagenase. Despite the presence of vitreous hemorrhages, which can possibly be avoided by optimization of the experimental set-up, we believe to have given the first step in the right direction towards nanotechnology-based enzymatic vitreolysis and to have opened doors for future research.
Collapse
Affiliation(s)
- An-Katrien Minnaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Van-Phuc Nguyen
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Aranit Harizaj
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Ilia Goemaere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Bart Van Puyvelde
- Laboratory of Pharmaceutical Biotechnology/ProGenTomics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Maarten Dhaenens
- Laboratory of Pharmaceutical Biotechnology/ProGenTomics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Félix Sauvage
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical Biotechnology/ProGenTomics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Kevin Braeckmans
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Centre for Advanced Light Microscopy, Ghent University, Ottergemsesteenweg 460, Ghent, 9000, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Yannis M Paulus
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
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
|
Titz B, Siebourg-Polster J, Bartolo F, Lavergne V, Jiang Z, Gayan J, Altay L, Enders P, Schmelzeisen C, Ippisch QT, Koss MJ, Ansari-Shahrezaei S, Garweg JG, Fauser S, Dieckmann A. Implications of Ocular Confounding Factors for Aqueous Humor Proteomic and Metabolomic Analyses in Retinal Diseases. Transl Vis Sci Technol 2024; 13:17. [PMID: 38913008 PMCID: PMC11205237 DOI: 10.1167/tvst.13.6.17] [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: 02/27/2024] [Accepted: 05/02/2024] [Indexed: 06/25/2024] Open
Abstract
Purpose To assess the impact of ocular confounding factors on aqueous humor (AH) proteomic and metabolomic analyses for retinal disease characterization. Methods This study recruited 138 subjects (eyes): 102 with neovascular age-related macular degeneration (nAMD), 18 with diabetic macular edema (DME), and 18 with cataract (control group). AH samples underwent analysis using Olink Target 96 proteomics and Metabolon's metabolomics platform Data analysis included correlation, differential abundance, and gene-set analysis. Results In total, 756 proteins and 408 metabolites were quantified in AH. Total AH protein concentration was notably higher in nAMD (3.2-fold) and DME (4.1-fold) compared to controls. Pseudophakic eyes showed higher total AH protein concentrations than phakic eyes (e.g., 1.6-fold in nAMD) and a specific protein signature indicative of matrix remodeling. Unexpectedly, pupil-dilating drugs containing phenylephrine/tropicamide increased several AH proteins, notably interleukin-6 (5.4-fold in nAMD). Correcting for these factors revealed functionally relevant protein correlation clusters and disease-relevant, differentially abundant proteins across the groups. Metabolomics analysis, for which the relevance of confounder adjustment was less apparent, suggested insufficiently controlled diabetes and chronic hyperglycemia in the DME group. Conclusions AH protein concentration, pseudophakia, and pupil dilation with phenylephrine/tropicamide are important confounding factors for AH protein analyses. When these factors are considered, AH analyses can more clearly reveal disease-relevant factors. Translational Relevance Considering AH protein concentration, lens status, and phenylephrine/tropicamide administration as confounders is crucial for accurate interpretation of AH protein data.
Collapse
Affiliation(s)
- Björn Titz
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Juliane Siebourg-Polster
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Francois Bartolo
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
- EFOR-CVO et Soladis, Champagne-au-Mont-d'Or, France
| | - Vincent Lavergne
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
- EFOR-CVO et Soladis, Basel, Switzerland
| | - Zhiwen Jiang
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Javier Gayan
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Lebriz Altay
- Department of Ophthalmology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Philip Enders
- Department of Ophthalmology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | | | | | | | | | - Justus Gerhard Garweg
- Berner Augenklinik, Bern, Switzerland
- Department of Ophthalmology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sascha Fauser
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Andreas Dieckmann
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| |
Collapse
|
5
|
Sahel DK, Goswami SG, Jatyan R, Tharmatt A, Singh V, Dalela M, Mohanty S, Mittal A, Ramalingam S, Chitkara D. cRGD-modified hybrid lipopolymeric nanoplexes for gene editing in the posterior segment of the eye. Int J Biol Macromol 2024; 271:132426. [PMID: 38820904 DOI: 10.1016/j.ijbiomac.2024.132426] [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: 10/01/2023] [Revised: 04/21/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024]
Abstract
Eye-related diseases, specifically retinal dystrophy (RD) conditions, are the leading cause of blindness worldwide. Gene addition, regulation, or editing could potentially treat such diseases through gene expression regulation. CRISPR/Cas9 gene editing is one of the most prominent and precise gene editing tools which could be employed to edit genes related to the dystrophic condition. However, CRISPR/Cas9 faces in vivo delivery challenges due to its high molecular weight, negative charge, prone to degradation in the presence of nucleases and proteases, poor cellular degradation, etc., which makes it challenging to adopt for therapeutic applications. We developed cRGD-modified lipopolymeric nanoplexes loaded with Cas9 RNPs with a particle size and zeta potential of 175 ± 20 nm and 2.15 ± 0.9 mV, respectively. The cRGD-modified lipopolymeric nanoplexes were stable for 194 h and able to transfect >70 % ARPE-19 and NIH3T3 cells with an Indel frequency of ~40 % for the VEGF-A gene. The cRGD-modified lipopolymeric nanoplexes found good vitreous mobility and could transfection retinal cells in vivo after 48 h of intravitreal injection in Wistar Rats. Moreover, in vivo VEGFA gene editing was ~10 % with minimal toxicities. Collectively, the cRGD-modified lipopolymeric nanoplexes were found to have extreme potential in delivering CRISPR/Cas9 RNPs payload to the retinal tissues for therapeutic applications.
Collapse
Affiliation(s)
- Deepak Kumar Sahel
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | | | - Reena Jatyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | - Abhay Tharmatt
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | - Vivek Singh
- Prof. Brien Holden Eye Research Center, Champalimaud Translational Centre for Eye Research, L.V. Prasad Eye Institute, Kallam Anji Reddy Campus, L V Prasad Marg, Hyderabad, India
| | - Manu Dalela
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India
| | | | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Pilani Campus, Vidya Vihar, Pilani, Rajasthan, India.
| |
Collapse
|
6
|
Carvalho SG, Haddad FF, Dos Santos AM, Scarim CB, Ferreira LMB, Meneguin AB, Chorilli M, Gremião MPD. Chitosan surface modification modulates the mucoadhesive, permeation and anti-angiogenic properties of gellan gum/bevacizumab nanoparticles. Int J Biol Macromol 2024; 263:130272. [PMID: 38373560 DOI: 10.1016/j.ijbiomac.2024.130272] [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: 09/20/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024]
Abstract
Bevacizumab (BVZ) was the first monoclonal antibody approved by the FDA and has shown an essential advance in the antitumor therapy of colorectal cancer (CRC), however, the systemic action of BVZ administered intravenously can trigger several adverse effects. The working hypothesis of the study was to promote the modulation of the mucoadhesion properties and permeability of the BVZ through the formation of nanoparticles (NPs) with gellan gum (GG) with subsequent surface modification with chitosan (CS). NPs comprising BVZ and GG were synthesized through polyelectrolyte complexation, yielding spherical nanosized particles with an average diameter of 264.0 ± 2.75 nm and 314.0 ± 0.01 nm, polydispersity index of 0.182 ± 0.01 e 0.288 ± 0.01, and encapsulation efficiency of 29.36 ± 0.67 e 60.35 ± 0.27 mV, for NPs without (NP_BVZ) and with surface modification (NP_BVZ + CS). The results showed a good ability of nanoparticles with surface modification to modulate the NPs biological properties.
Collapse
Affiliation(s)
- Suzana Gonçalves Carvalho
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Felipe Falcão Haddad
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Aline Martins Dos Santos
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Cauê Benito Scarim
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Leonardo Miziara Barboza Ferreira
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil
| | - Andréia Bagliotti Meneguin
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Marlus Chorilli
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| | - Maria Palmira Daflon Gremião
- Department of Drugs and Pharmaceutics, School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800-903 Araraquara, SP, Brazil.
| |
Collapse
|
7
|
Rupenthal ID, Agarwal P. Progress in Ocular Drug Delivery: Challenges and Constraints. Handb Exp Pharmacol 2024; 284:267-288. [PMID: 37620616 DOI: 10.1007/164_2023_693] [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] [Indexed: 08/26/2023]
Abstract
The eye has several dynamic and static barriers in place to limit the entry of foreign substances including therapeutics. As such, efficient drug delivery, especially to posterior segment tissues, has been challenging. This chapter describes the anatomical and physiological challenges associated with ocular drug delivery before discussing constraints with regard to formulation parameters. Finally, it gives an overview of advanced drug delivery technologies with a specific focus on recently marketed and late-stage clinical trial products.
Collapse
Affiliation(s)
- Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Priyanka Agarwal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
8
|
Wilson S, Siebourg-Polster J, Titz B, Jiang Z, Bartolo F, Lavergne V, Gayán J, Garweg JG, Fauser S, Dieckmann A. Correlation of Aqueous, Vitreous, and Serum Protein Levels in Patients With Retinal Diseases. Transl Vis Sci Technol 2023; 12:9. [PMID: 37930665 PMCID: PMC10629536 DOI: 10.1167/tvst.12.11.9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
Purpose To further establish aqueous humor (AH) as a clinically suitable source of protein biomarkers in retinal diseases by evaluating the correlation of a large panel of proteins between AH, vitreous humor (VH), and serum (SE). Methods We enrolled 60 subjects (eyes) with various non-infectious retinal diseases. AH, VH, and SE proteins were analyzed using the Olink Target 96 platform (1196 protein assays in total). We compared these three matrices in terms of quantification overlap, principal component analysis, and correlation. Results In the AH, VH, and SE samples, 841, 917, and 1133 proteins, respectively, were consistently quantified above the limit of detection in more than 30% of patients. AH and VH shared 812 of these proteins. AH and VH samples overlapped along principal component 1, but SE samples were distinct. We identified 490 proteins with significant (false discovery rate [FDR]-adjusted P < 0.05) and relevant correlations (correlation coefficient > 0.5) between AH and VH, compared to only 33 and 40 proteins for VH and SE and for AH and SE, respectively. Conclusions Due to a close correlation between protein concentrations in the AH and VH and a clear difference from the SE, AH has the potential to serve as a substitute for VH and may hold significance in identifying protein biomarkers and novel targets related to retinal diseases. Translational Relevance This study further supports AH as a clinically suitable source of protein biomarkers in retinal diseases. In addition, the identified AH and VH correlations can inform the selection of protein biomarker candidates in future translational research.
Collapse
Affiliation(s)
- Sabine Wilson
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | - Bjoern Titz
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Zhiwen Jiang
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Francois Bartolo
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- EFOR-CVO et Soladis, Champagne-au-Mont-d'Or, France
| | - Vincent Lavergne
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- EFOR-CVO et Soladis, Basel, Switzerland
| | - Javier Gayán
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Justus G. Garweg
- Berner Augenklinik, Bern, Switzerland
- Department of Ophthalmology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sascha Fauser
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andreas Dieckmann
- Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
9
|
Eriksen AZ, Melander F, Eriksen GDM, Kempen PJ, Kjaer A, Andresen TL, Urquhart AJ. Active Transport and Ocular Distribution of Intravitreally Injected Liposomes. Transl Vis Sci Technol 2023; 12:20. [PMID: 37615641 PMCID: PMC10461645 DOI: 10.1167/tvst.12.8.20] [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: 01/12/2022] [Accepted: 07/10/2023] [Indexed: 08/25/2023] Open
Abstract
Purpose Drug delivery to the retina remains a challenge due to ocular barriers and fast clearing mechanisms. Nanocarrier drug delivery systems (NDDSs) hold the promise of prolonging intraocular retention times and increasing drug concentrations in the retina. Methods Anionic and cationic PEGylated liposomes, loaded with oxaliplatin (OxPt) to be used as trace element, were prepared from dry lipid powders. The differently charged liposomes were intravitreally injected in C57BL/6JrJ mice; eyes were harvested 2 hours and 24 hours post-injection. To investigate active transport mechanisms in the eye, a subset of mice were pre-injected with chloroquine before injection with cationic liposomes. Eyes were dissected and the distribution of OxPt in different tissues were quantified by inductively coupled plasma mass spectrometry (ICP-MS). Results Both liposome formulations enhanced the retention time of OxPt in the vitreous over free OxPt. Surprisingly, when formulated in cationic liposomes, OxPt translocated through the retina and accumulated in the RPE-sclera. Pre-injection with chloroquine inhibited the transport of liposomal OxPt from the vitreous to the RPE-sclera. Conclusions We show that liposomes can enhance the retention time of small molecular drugs in the vitreous and that active transport mechanisms are involved in the trans retinal transport of NDDS after intravitreal injections. Translational Relevance These results highlight the need for understanding the dynamics of ocular transport mechanisms in living eyes when designing NDDS with the back of the eye as the target. Active transport of nanocarriers through the retina will limit the drug concentration in the neuronal retina but might be exploited for targeting the RPE.
Collapse
Affiliation(s)
- Anne Zebitz Eriksen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Fredrik Melander
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | - Paul Joseph Kempen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- National Center for Nano Fabrication and Characterization, Technical University of Denmark, Lyngby, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET & Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | |
Collapse
|
10
|
Supe S, Upadhya A, Tripathi S, Dighe V, Singh K. Liposome-polyethylenimine complexes for the effective delivery of HuR siRNA in the treatment of diabetic retinopathy. Drug Deliv Transl Res 2023; 13:1675-1698. [PMID: 36630075 DOI: 10.1007/s13346-022-01281-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 01/12/2023]
Abstract
Diabetic retinopathy (DR) is a vision-impairing complication of diabetes, damaging the retinal microcirculatory system. Overexpression of VEGF (vascular endothelial growth factor) is implicated in the pathogenesis of DR. Human antigen R (HuR) is an RNA-binding protein that favorably regulates VEGF protein expression by binding to VEGF-encoding mRNA. Downregulating HuR via RNA interference strategies using small interfering RNAs (siRNAs) may constitute a novel therapeutic method for preventing VEGF protein overexpression in DR. Delivery of siRNAs to the cellular cytoplasm can be facilitated by cationic peptides or polymers and lipids. In this study, a cationic polymer (polyethylenimine (PEI)) and lipid nanoparticles (liposomes) were co-formulated with siRNA to form lipopolyplexes (LPPs) for the delivery of HuR siRNA. LPPs-siRNA were analyzed for size, zeta potential, serum stability, RNase stability, heparin stability, toxicity, and siRNA encapsulation efficiency. Cellular uptake, downregulation of the target HuR (mRNA and protein), and associated VEGF protein were used to demonstrate the biological efficacy of the LPPs-HuR siRNA, in vitro (human ARPE-19 cells), and in vivo (Wistar rats). In vivo efficacy study was performed by injecting LPPs-HuR siRNA formulations into the eye of streptozotocin (STZ)-induced diabetic rats after the development of retinopathy. Our findings demonstrated that high retinal HuR and VEGF levels observed in the eyes of untreated STZ rats were lowered after LPPs-HuR siRNA administration. Our observations indicate that intravitreal treatment with HuR siRNA is a promising option for DR using LPPs as delivery agents.
Collapse
Affiliation(s)
- Shibani Supe
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, 400056, India
| | - Archana Upadhya
- Humera Khan College of Pharmacy, HK College Campus, Oshiwara, Jogeshwari (West), Mumbai, Maharashtra, 400102, India
| | - Santosh Tripathi
- Bombay Veterinary College, Sindhu Nagar, Parel Village, Parel, Mumbai, Maharashtra, 400012, India
| | - Vikas Dighe
- National Centre for Preclinical Reproductive and Genetic Toxicology, ICMR-National Institute for Research in Reproductive and Child Health, J.M.Street, Parel, Mumbai, Maharashtra, 400012, India.
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
11
|
Zashikhina N, Gladnev S, Sharoyko V, Korzhikov-Vlakh V, Korzhikova-Vlakh E, Tennikova T. Synthesis and Characterization of Nanoparticle-Based Dexamethasone-Polypeptide Conjugates as Potential Intravitreal Delivery Systems. Int J Mol Sci 2023; 24:ijms24043702. [PMID: 36835114 PMCID: PMC9962198 DOI: 10.3390/ijms24043702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
The use of dexamethasone for eye disease treatment is limited by its low solubility, bioavailability, and rapid elimination when applied topically. The covalent conjugation of dexamethasone with polymeric carriers is a promising strategy to overcome existing drawbacks. In this work, amphiphilic polypeptides capable of self-assembly into nanoparticles were proposed as potential delivery systems for intravitreal delivery. The nanoparticles were prepared and characterized using poly(L-glutamic acid-co-D-phenylalanine) and poly(L-lysine-co-D/L-phenylalanine) as well as poly(L-lysine-co-D/L-phenylalanine) covered with heparin. The critical association concentration for the polypeptides obtained was in the 4.2-9.4 μg/mL range. The hydrodynamic size of the formed nanoparticles was between 90 and 210 nm, and they had an index of polydispersity between 0.08 and 0.27 and an absolute zeta-potential value between 20 and 45 mV. The ability of nanoparticles to migrate in the vitreous humor was examined using intact porcine vitreous. Conjugation of DEX with polypeptides was performed by additional succinylation of DEX and activation of carboxyl groups introduced to react with primary amines in polypeptides. The structures of all intermediate and final compounds were verified by 1H NMR spectroscopy. The amount of conjugated DEX can be varied from 6 to 220 µg/mg of polymer. The hydrodynamic diameter of the nanoparticle-based conjugates was increased to 200-370 nm, depending on the polymer sample and drug loading. The release of DEX from the conjugates due to hydrolysis of the ester bond between DEX and the succinyl moiety was studied both in a buffer medium and a vitreous/buffer mixture (50/50, v/v). As expected, the release in the vitreous medium was faster. However, the release rate could be controlled in the range of 96-192 h by varying the polymer composition. In addition, several mathematical models were used to assess the release profiles and figure out how DEX is released.
Collapse
Affiliation(s)
- Natalia Zashikhina
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg 199004, Russia
| | - Sergei Gladnev
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg 198504, Russia
| | - Vladimir Sharoyko
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg 198504, Russia
- Department of General and Bioorganic Chemistry, Pavlov First Saint-Petersburg State Medical University, L’va Tolstogo str. 6-8, St. Petersburg 197022, Russia
| | - Viktor Korzhikov-Vlakh
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg 198504, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg 199004, Russia
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg 198504, Russia
- Correspondence:
| | - Tatiana Tennikova
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg 198504, Russia
| |
Collapse
|
12
|
Peynshaert K, Devoldere J, De Smedt S, Remaut K. Every nano-step counts: a critical reflection on do's and don'ts in researching nanomedicines for retinal gene therapy. Expert Opin Drug Deliv 2023; 20:259-271. [PMID: 36630275 DOI: 10.1080/17425247.2023.2167979] [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: 01/12/2023]
Abstract
INTRODUCTION Retinal disease affects millions of people worldwide, generating a massive social and economic burden. Current clinical trials for retinal diseases are dominated by gene augmentation therapies delivered with recombinant viruses as key players. As an alternative, nanoparticles hold great promise for the delivery of nucleic acid therapeutics as well. Nevertheless, despite numerous attempts, 'nano' is in practice not as successful as aspired and major breakthroughs in retinal gene therapy applying nanomaterials are yet to be seen. AREAS COVERED In this review, we summarize the advantages of nanomaterials and give an overview of nanoparticles designed for retinal nucleic acid delivery up to now. We furthermore critically reflect on the predominant issues that currently limit nano to progress to the clinic, where faulty study design and the absence of representative models play key roles. EXPERT OPINION Since the current approach of in vitro - in vivo experimentation is highly inefficient and creates misinformation, we advocate for a more prominent role for ex vivo testing early on in nanoparticle research. In addition, we elaborate on several concepts, including systematic studies and open science, which could aid in pushing the field of nanomedicine beyond the preclinical stage.
Collapse
Affiliation(s)
- Karen Peynshaert
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Belgium Belgium.,Ghent Research Group on Nanomedicines, Ghent University, Belgium Belgium
| | - Joke Devoldere
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Belgium Belgium.,Ghent Research Group on Nanomedicines, Ghent University, Belgium Belgium
| | - Stefaan De Smedt
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Belgium Belgium.,Ghent Research Group on Nanomedicines, Ghent University, Belgium Belgium
| | - Katrien Remaut
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Belgium Belgium.,Ghent Research Group on Nanomedicines, Ghent University, Belgium Belgium
| |
Collapse
|
13
|
Photodisruption of the Inner Limiting Membrane: Exploring ICG Loaded Nanoparticles as Photosensitizers. Pharmaceutics 2022; 14:pharmaceutics14081716. [PMID: 36015342 PMCID: PMC9416162 DOI: 10.3390/pharmaceutics14081716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
The inner limiting membrane (ILM) represents a major bottleneck hampering efficient drug delivery to the retina after intravitreal injection. To overcome this barrier, we intend to perforate the ILM by use of a light-based approach which relies on the creation of vapor nanobubbles (VNBs) when irradiating photosensitizers with high intensity laser pulses. Upon collapse of these VNBs, mechanical effects can disrupt biological structures. As a photosensitizer, we explore indocyanine green (ICG) loaded nanoparticles (NPs) specifically designed for our application. In light of this, ICG liposomes and PLGA ICG NPs were characterized in terms of physicochemical properties, ICG incorporation and VNB formation. ICG liposomes were found to encapsulate significantly higher amounts of ICG compared to PLGA ICG NPs which is reflected in their VNB creating capacity. Since only ICG liposomes were able to induce VNB generation, this class of NPs was further investigated on retinal explants. Here, application of ICG liposomes followed by laser treatment resulted in subtle disruption effects at the ILM where zones of fully ablated ILM were alternated by intact regions. As the interaction between the ICG liposomes and ILM might be insufficient, active targeting strategies or other NP designs might improve the concept to a further extent.
Collapse
|
14
|
Yeast-produced fructosamine-3-kinase retains mobility after ex vivo intravitreal injection in human and bovine eyes as determined by Fluorescence Correlation Spectroscopy. Int J Pharm 2022; 621:121772. [PMID: 35487399 DOI: 10.1016/j.ijpharm.2022.121772] [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: 02/02/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/20/2022]
Abstract
Globally, over 2 billion people suffer from vision impairment. Despite complex multifactorial etiology, advanced glycation end products are involved in the pathogenesis of many causative age- and diabetes-related eye diseases. Deglycating enzyme fructosamine-3-kinase (FN3K) was recently proposed as a potential therapeutic, but for further biopharmaceutical development, knowledge on its manufacturability and stability and mobility in the vitreous fluid of the eye is indispensable. We evaluated recombinant production of FN3K in two host systems, and its diffusion behavior in both bovine and human vitreous. Compared to Escherichia coli, intracellular production in Pichia pastoris yielded more and higher purity FN3K. The yeast-produced enzyme was used in a first attempt to use fluorescence correlation spectroscopy to study protein mobility in non-sonicated bovine vitreous, human vitreous, and intact bovine eyes. It was demonstrated that FN3K retained mobility upon intravitreal injection, although a certain delay in diffusion was observed. Alkylation of free cysteines was tolerated both in terms of enzymatic activity and vitreous diffusion. Ex vivo diffusion data gathered and the availability of yeast-produced high purity enzyme now clear the path for in vivo pharmacokinetics studies of FN3K.
Collapse
|
15
|
Berger S, Berger M, Bantz C, Maskos M, Wagner E. Performance of nanoparticles for biomedical applications: The in vitro/ in vivo discrepancy. BIOPHYSICS REVIEWS 2022; 3:011303. [PMID: 38505225 PMCID: PMC10903387 DOI: 10.1063/5.0073494] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/04/2022] [Indexed: 03/21/2024]
Abstract
Nanomedicine has a great potential to revolutionize the therapeutic landscape. However, up-to-date results obtained from in vitro experiments predict the in vivo performance of nanoparticles weakly or not at all. There is a need for in vitro experiments that better resemble the in vivo reality. As a result, animal experiments can be reduced, and potent in vivo candidates will not be missed. It is important to gain a deeper knowledge about nanoparticle characteristics in physiological environment. In this context, the protein corona plays a crucial role. Its formation process including driving forces, kinetics, and influencing factors has to be explored in more detail. There exist different methods for the investigation of the protein corona and its impact on physico-chemical and biological properties of nanoparticles, which are compiled and critically reflected in this review article. The obtained information about the protein corona can be exploited to optimize nanoparticles for in vivo application. Still the translation from in vitro to in vivo remains challenging. Functional in vitro screening under physiological conditions such as in full serum, in 3D multicellular spheroids/organoids, or under flow conditions is recommended. Innovative in vivo screening using barcoded nanoparticles can simultaneously test more than hundred samples regarding biodistribution and functional delivery within a single mouse.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig–Maximilians-Universität (LMU) Munich, Butenandtstr. 5-13, D-81377 Munich, Germany
| | - Martin Berger
- Department of Chemistry, Johannes Gutenberg-Universität Mainz, Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Christoph Bantz
- Fraunhofer Institute for Microengineering and Microsystems IMM, Carl-Zeiss-Str. 18-20, D-55129 Mainz, Germany
| | | | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig–Maximilians-Universität (LMU) Munich, Butenandtstr. 5-13, D-81377 Munich, Germany
| |
Collapse
|
16
|
A core-shell nanoplatform as a nonviral vector for targeted delivery of genes to the retina. Acta Biomater 2021; 134:605-620. [PMID: 34329781 DOI: 10.1016/j.actbio.2021.07.053] [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] [Received: 04/22/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 01/05/2023]
Abstract
Retinal diseases, including age-related macular degeneration (AMD), are a major cause of blindness. Efficient delivery of therapeutic genes to retinal cells to treat retinal disease is a formidable challenge. In this study, we developed a core-shell nanoplatform composed of a core and two external layers for targeted delivery of the gene to the retina. The inner core was composed of amino acid-functionalized dendrimers and a nuclear localization signal (NLS) for DNA complexation, nuclear transport and efficient transfection. The inner core was coated in a lipid bilayer that comprised pH-sensitive lipids as the inner shell layer. Hyaluronic acid (HA)-1,2-dioleoylphosphatidylethanolamine (DOPE) as the outermost shell layer was used for retinal cell targeting. This core-shell nanoplatform was developed so that the mobility in the vitreous body of these negatively charged carriers would not be affected by their surface charge, allowing diffusion into the retina, uptake into the retinal cells via CD44-mediated internalization, and finally transport into the nucleus by the NLS. The designed nanoparticles showed safety both in vitro and in vivo and inhibited the expression of VEGF under hypoxia-mimicking conditions. In vitro angiogenesis assays exhibited significant inhibitory effects on cell migration and tube formation. The in vivo assays indicated that this nanoplatform could be delivered to the retina. Taken together, this nanoplatform has the potential to transfer gene material into the retina for the treatment of retinal diseases, including AMD. STATEMENT OF SIGNIFICANCE: It remains a challenge to develop an efficient nonviral vector for gene therapy, especially retinal gene therapy. Various barriers exist in gene delivery and the unique ocular environment, making gene delivery to the retina difficult. In this study, we designed a negatively charged core-shell nanoplatform (HD-NPPND) for the targeted delivery of gene to the retina. The developed nanoplatform possessed excellent transfection efficiency and safety both in vitro and in vivo. It efficiently delivered a gene to the retina. The results of this study suggested that this core-shell nanoplatform has the potential to deliver genes to the retina to treat retinal diseases, including age-related macular degeneration (AMD).
Collapse
|
17
|
Synthesis and Characterization of Novel Succinyl Chitosan-Dexamethasone Conjugates for Potential Intravitreal Dexamethasone Delivery. Int J Mol Sci 2021; 22:ijms222010960. [PMID: 34681619 PMCID: PMC8535746 DOI: 10.3390/ijms222010960] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/25/2022] Open
Abstract
The development of intravitreal glucocorticoid delivery systems is a current global challenge for the treatment of inflammatory diseases of the posterior segment of the eye. The main advantages of these systems are that they can overcome anatomical and physiological ophthalmic barriers and increase local bioavailability while prolonging and controlling drug release over several months to improve the safety and effectiveness of glucocorticoid therapy. One approach to the development of optimal delivery systems for intravitreal injections is the conjugation of low-molecular-weight drugs with natural polymers to prevent their rapid elimination and provide targeted and controlled release. This study focuses on the development of a procedure for a two-step synthesis of dexamethasone (DEX) conjugates based on the natural polysaccharide chitosan (CS). We first used carbodiimide chemistry to conjugate DEX to CS via a succinyl linker, and we then modified the obtained systems with succinic anhydride to impart a negative ζ-potential to the polymer particle surface. The resulting polysaccharide carriers had a degree of substitution with DEX moieties of 2–4%, a DEX content of 50–85 μg/mg, and a degree of succinylation of 64–68%. The size of the obtained particles was 400–1100 nm, and the ζ-potential was −30 to −33 mV. In vitro release studies at pH 7.4 showed slow hydrolysis of the amide and ester bonds in the synthesized systems, with a total release of 8–10% for both DEX and succinyl dexamethasone (SucDEX) after 1 month. The developed conjugates showed a significant anti-inflammatory effect in TNFα-induced and LPS-induced inflammation models, suppressing CD54 expression in THP-1 cells by 2- and 4-fold, respectively. Thus, these novel succinyl chitosan-dexamethasone (SucCS-DEX) conjugates are promising ophthalmic carriers for intravitreal delivery.
Collapse
|
18
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
19
|
Plaza-Oliver M, Cano EL, Arroyo-Jimenez MM, Gámez M, Lozano-López MV, Santander-Ortega MJ. Taking Particle Tracking into Practice by Novel Software and Screening Approach: Case-Study of Oral Lipid Nanocarriers. Pharmaceutics 2021; 13:370. [PMID: 33802226 PMCID: PMC8001040 DOI: 10.3390/pharmaceutics13030370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
The success on the design of new oral nanocarriers greatly depends on the identification of the best physicochemical properties that would allow their diffusion across the mucus layer that protects the intestinal epithelium. In this context, particle tracking (PT) has arisen in the pharmaceutical field as an excellent tool to evaluate the diffusion of individual particles across the intestinal mucus. In PT, the trajectories of individual particles are characterized by the mean square displacement (MSD), which is used to calculate the coefficient of diffusion (D) and the anomalous diffusion parameter (α) as MSD=4Dτα. Unfortunately, there is no stablished criteria to evaluate the goodness-of-fit of the experimental data to the mathematical model. This work shows that the commonly used R2 parameter may lead to an overestimation of the diffusion capacity of oral nanocarriers. We propose a screening approach based on a combination of R2 with further statistical parameters. We have analyzed the effect of this approach to study the intestinal mucodiffusion of lipid oral nanocarriers, compared to the conventional screening approach. Last, we have developed software able to perform the whole PT analysis in a time-saving, user-friendly, and rational fashion.
Collapse
Affiliation(s)
- María Plaza-Oliver
- Cellular Neurobiology and Molecular Chemistry of the Central Nervous System Group, Faculty of Pharmacy, University of Castilla-La Mancha (UCLM), 02071 Albacete, Spain; (M.P.-O.); (M.M.A.-J.); (M.V.L.-L.)
- Regional Centre of Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Emilio L. Cano
- Quantitative Methods and Socio-economic Development Group, Institute for Regional Development (IDR), University of Castilla-La Mancha (UCLM), 02006 Albacete, Spain;
- Data Science laboratory, Rey Juan Carlos University, 28933 Madrid, Spain
| | - María Mar Arroyo-Jimenez
- Cellular Neurobiology and Molecular Chemistry of the Central Nervous System Group, Faculty of Pharmacy, University of Castilla-La Mancha (UCLM), 02071 Albacete, Spain; (M.P.-O.); (M.M.A.-J.); (M.V.L.-L.)
- Regional Centre of Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Matías Gámez
- Quantitative Methods and Socio-economic Development Group, Institute for Regional Development (IDR), University of Castilla-La Mancha (UCLM), 02006 Albacete, Spain;
| | - María Victoria Lozano-López
- Cellular Neurobiology and Molecular Chemistry of the Central Nervous System Group, Faculty of Pharmacy, University of Castilla-La Mancha (UCLM), 02071 Albacete, Spain; (M.P.-O.); (M.M.A.-J.); (M.V.L.-L.)
- Regional Centre of Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| | - Manuel J. Santander-Ortega
- Cellular Neurobiology and Molecular Chemistry of the Central Nervous System Group, Faculty of Pharmacy, University of Castilla-La Mancha (UCLM), 02071 Albacete, Spain; (M.P.-O.); (M.M.A.-J.); (M.V.L.-L.)
- Regional Centre of Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), 02008 Albacete, Spain
| |
Collapse
|
20
|
Balasso A, Subrizi A, Salmaso S, Mastrotto F, Garofalo M, Tang M, Chen M, Xu H, Urtti A, Caliceti P. Screening of chemical linkers for development of pullulan bioconjugates for intravitreal ocular applications. Eur J Pharm Sci 2021; 161:105785. [PMID: 33667663 DOI: 10.1016/j.ejps.2021.105785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
The treatment of posterior segment disorders of the eye requires therapeutic strategies to achieve drug effects over prolonged times. Innovative colloidal delivery systems can be designed to deliver drugs to the retina and prolong their intravitreal permanence. In order to exploit pullulan (Pull) as polymeric drug carrier for intravitreal drug delivery, derivatives of hydrophobic model molecule rhodamine B (RhB) were conjugated to the pullulan backbone through linkers with different stability, namely ether (Et), hydrazone (Hy) or ester (Es) bond to obtain Pull-Et-RhB, Pull-Hy-RhB and Pull-Es-RhB, respectively. Dynamic light scattering and transmission electron microscopy analyses showed that the polymer conjugates self-assembled into 20-25 nm particles. Pull-Et-RhB was fairly stable at all tested pH values. At the vitreal pH of 7.4, 50% of RhB was released from Pull-Hy-RhB and Pull-Es-RhB in 11 and 6 days, respectively. At endosomal pH (5.5), 50% of RhB was released from Pull-Hy-RhB and Pull-Es-RhB in 4 and 1 days, respectively. Multiple particle tracking analyses in ex vivo porcine eye model showed that the diffusivity of the bioconjugates in the vitreous was about 103 times lower than in water. Flow cytometry and confocal microscopy analyses showed that bioconjugates are remarkably taken up by the retinal RPE cells. In vivo studies showed that after intravitreal injection to mice, the bioconjugates localize in the ganglion cell layer and in the inner and outer plexiform layers. Pull-Hy-RhB particles were detected also inside the retinal blood vessels. These results demonstrate that pullulan with tailored linkers for drug conjugation is a promising vehicle for long-acting intravitreal injectables that are capable to permeate to the retina.
Collapse
Affiliation(s)
- Anna Balasso
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy
| | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
| | - Stefano Salmaso
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy
| | - Francesca Mastrotto
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy
| | - Mariangela Garofalo
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy
| | - Miao Tang
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland; Drug Research Program, Division of Pharmaceutical Biosciences, University of Helsinki, POB 56, 00014 University of Helsinki, Finland; Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya Embankment, 199034 St. Petersburg, Russian Federation.
| | - Paolo Caliceti
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy.
| |
Collapse
|
21
|
Pishavar E, Luo H, Bolander J, Atala A, Ramakrishna S. Nanocarriers, Progenitor Cells, Combinational Approaches, and New Insights on the Retinal Therapy. Int J Mol Sci 2021; 22:1776. [PMID: 33579019 PMCID: PMC7916765 DOI: 10.3390/ijms22041776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Progenitor cells derived from the retinal pigment epithelium (RPECs) have shown promise as therapeutic approaches to degenerative retinal disorders including diabetic retinopathy, age-related macular degeneration and Stargardt disease. However, the degeneration of Bruch's membrane (BM), the natural substrate for the RPE, has been identified as one of the major limitations for utilizing RPECs. This degeneration leads to decreased support, survival and integration of the transplanted RPECs. It has been proposed that the generation of organized structures of nanofibers, in an attempt to mimic the natural retinal extracellular matrix (ECM) and its unique characteristics, could be utilized to overcome these limitations. Furthermore, nanoparticles could be incorporated to provide a platform for improved drug delivery and sustained release of molecules over several months to years. In addition, the incorporation of tissue-specific genes and stem cells into the nanostructures increased the stability and enhanced transfection efficiency of gene/drug to the posterior segment of the eye. This review discusses available drug delivery systems and combination therapies together with challenges associated with each approach. As the last step, we discuss the application of nanofibrous scaffolds for the implantation of RPE progenitor cells with the aim to enhance cell adhesion and support a functionally polarized RPE monolayer.
Collapse
Affiliation(s)
- Elham Pishavar
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91735, Iran;
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Hongrong Luo
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, China;
| | - Johanna Bolander
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Antony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology, National University of Singapore, Singapore 117581, Singapore
| |
Collapse
|
22
|
Tavakoli S, Kari OK, Turunen T, Lajunen T, Schmitt M, Lehtinen J, Tasaka F, Parkkila P, Ndika J, Viitala T, Alenius H, Urtti A, Subrizi A. Diffusion and Protein Corona Formation of Lipid-Based Nanoparticles in the Vitreous Humor: Profiling and Pharmacokinetic Considerations. Mol Pharm 2021; 18:699-713. [PMID: 32584047 PMCID: PMC7856631 DOI: 10.1021/acs.molpharmaceut.0c00411] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/18/2020] [Accepted: 06/25/2020] [Indexed: 12/30/2022]
Abstract
The vitreous humor is the first barrier encountered by intravitreally injected nanoparticles. Lipid-based nanoparticles in the vitreous are studied by evaluating their diffusion with single-particle tracking technology and by characterizing their protein coronae with surface plasmon resonance and high-resolution proteomics. Single-particle tracking results indicate that the vitreal mobility of the formulations is dependent on their charge. Anionic and neutral formulations are mobile, whereas larger (>200 nm) neutral particles have restricted diffusion, and cationic particles are immobilized in the vitreous. PEGylation increases the mobility of cationic and larger neutral formulations but does not affect anionic and smaller neutral particles. Convection has a significant role in the pharmacokinetics of nanoparticles, whereas diffusion drives the transport of antibodies. Surface plasmon resonance studies determine that the vitreal corona of anionic formulations is sparse. Proteomics data reveals 76 differentially abundant proteins, whose enrichment is specific to either the hard or the soft corona. PEGylation does not affect protein enrichment. This suggests that protein-specific rather than formulation-specific factors are drivers of protein adsorption on nanoparticles in the vitreous. In summary, our findings contribute to understanding the pharmacokinetics of nanoparticles in the vitreous and help advance the development of nanoparticle-based treatments for eye diseases.
Collapse
Affiliation(s)
- Shirin Tavakoli
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Otto Kalevi Kari
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Tiina Turunen
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Tatu Lajunen
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Mechthild Schmitt
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Julia Lehtinen
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Fumitaka Tasaka
- Pharmaceutics
& Pharmacology Department, Global R&D, Santen Pharmaceutical
Co., Ltd., 8916-16 Takayama-cho, Ikoma, Nara 630-0101, Japan
| | - Petteri Parkkila
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Joseph Ndika
- Human
Microbiome Research, Faculty of Medicine, University of Helsinki, P.O. Box 21, 00290 Helsinki, Finland
| | - Tapani Viitala
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
| | - Harri Alenius
- Human
Microbiome Research, Faculty of Medicine, University of Helsinki, P.O. Box 21, 00290 Helsinki, Finland
- Institute
of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Arto Urtti
- Drug
Research Program, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014, Helsinki, Finland
- Institute
of Chemistry, St. Petersburg State University, Petergof, Universitetskii pr. 26, 198504 St. Petersburg, Russia
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Astrid Subrizi
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| |
Collapse
|
23
|
Rescue the retina after the ischemic injury by polymer-mediated intracellular superoxide dismutase delivery. Biomaterials 2020; 268:120600. [PMID: 33360507 DOI: 10.1016/j.biomaterials.2020.120600] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/14/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
Oxidative stress is a hallmark of the pathophysiogenesis of retinal ischemia. The direct delivery of antioxidant enzymes such as superoxide dismutase (SOD) into retinal cells provides a promising option for the down-regulation of oxidative stress in retinal ischemia, however, efficient intracellular protein delivery remains a major challenge for this application. Here, a boronic acid-rich polymer was used for the intracellular delivery of SOD both in vitro and in vivo. The polymer assembled with SOD into uniform nanoparticles with high binding affinity, and transported the cargo protein into several cell lines with maintained bioactivity and low cytotoxicity. We investigated the intraocular biodistribution, therapeutic efficacy and safety of the SOD nanoformulation in a retinal ischemia/reperfusion (I/R) injury model. After intravitreal injection, the nanoparticles rapidly diffused through the vitreous and penetrated into retinal ganglion cells (RGCs). Compared to free SOD, the nanoformulation exhibited much enhanced therapeutic efficacy with reduced RGC apoptosis and protected retinal function. Enzymatic results confirmed that the SOD nanoformulation reduced malondialdehyde expression and increased glutathione level in the ocular tissues, and thereby down-regulated oxidative stress and prevented RGC loss. Overall, this work offers a new therapeutic option for the treatment of retinal ischemic disorders by direct delivery of antioxidant proteins.
Collapse
|
24
|
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
|
25
|
Tavakoli S, Peynshaert K, Lajunen T, Devoldere J, Del Amo EM, Ruponen M, De Smedt SC, Remaut K, Urtti A. Ocular barriers to retinal delivery of intravitreal liposomes: Impact of vitreoretinal interface. J Control Release 2020; 328:952-961. [PMID: 33091527 DOI: 10.1016/j.jconrel.2020.10.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/24/2022]
Abstract
Drug delivery to the posterior segment of the eye is challenging due to several anatomical and physiological barriers. Thus, there is a need for prolonged action and targeted drug delivery to treat retinal diseases. Intravitreal injections avoid anterior eye barriers, but the vitreoretinal interface and inner limiting membrane (ILM) may prevent access of drug delivery systems to the retina. Existing data on retinal permeation of intravitreal nanoparticles are sparse and probably misleading due to the inter-species differences of retinal structures in rodents and humans. To bridge this gap, retinal permeation of light-activated liposomes was studied in an ex vivo bovine explant system that simulates the structure of vitreoretinal interface and intact ILM. Our findings indicate that the particle size plays a significant role in determining the retinal penetration as the liposomes of >100 nm sized failed to overcome the ILM and could not permeate into the retina. In addition, our results demonstrate the impact of surface charge and PEG-coating on retinal penetration. Small (≈ 50 nm) anionic liposomes with PEG coating showed the most extensive distribution and cellular localization in the retina. In summary, this study extends understanding of ocular barriers, and provides valuable information to augment design of retinal drug delivery systems.
Collapse
Affiliation(s)
- Shirin Tavakoli
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Tatu Lajunen
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Science, Tokyo University of Pharmacy & Life Sciences, 1432-1 Hachioji, 192-0392 Tokyo, Japan
| | - Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Eva M Del Amo
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marika Ruponen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Arto Urtti
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland; Institute of Chemistry, St Petersburg State University, Petergoff, St Petersburg, Russian Federation
| |
Collapse
|
26
|
Zeng Y, Boyd R, Bartoe J, Wiley HE, Marangoni D, Wei LL, Sieving PA. "Para-retinal" Vector Administration into the Deep Vitreous Enhances Retinal Transgene Expression. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:422-427. [PMID: 32695844 PMCID: PMC7363691 DOI: 10.1016/j.omtm.2020.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/18/2020] [Indexed: 01/22/2023]
Abstract
Intravitreal administration for human adeno-associated vector (AAV) delivery is easier and less traumatic to ocular tissues than subretinal injection, but it gives limited retinal transduction. AAV vectors are large (about 4,000 kDa) compared with most intraocular drugs, such as ranibizumab (48 kDa), and the large size impedes diffusion to reach the retina from the usual injection site in the anterior/mid-vitreous. Intuitively, a preferred placement for the vector would be deep in the vitreous near the retina, which we term “para-retinal” delivery. We explored the consequences of para-retinal intravitreal delivery in the rabbit eye and in non-human primate (NHP) eye. 1 h after para-retinal administration in the rabbit eye, the vector concentration near the retina remained four times greater than in the anterior vitreous, indicating limited vector diffusion through the gelatinous vitreous matrix. In NHP, para-retinal placement showed greater transduction in the fovea than vector applied in the mid-vitreous. More efficient retinal delivery translates to using lower vector doses, with reduced risk of ocular inflammatory exposure. These results indicate that para-retinal delivery yields more effective vector concentration near the retina, thereby increasing the potential for better retinal transduction in human clinical application.
Collapse
Affiliation(s)
- Yong Zeng
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ryan Boyd
- Charles River Laboratories, Matawan, MI, USA
| | | | - Henry E Wiley
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dario Marangoni
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa L Wei
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul A Sieving
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA.,Center for Ocular Regenerative Therapy; Department of Ophthalmology, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
27
|
Kim HM, Ha S, Hong HK, Hwang Y, Kim P, Yang E, Chung JY, Park S, Park YJ, Park KH, Kim H, Woo SJ. Intraocular Distribution and Kinetics of Intravitreally Injected Antibodies and Nanoparticles in Rabbit Eyes. Transl Vis Sci Technol 2020; 9:20. [PMID: 32821517 PMCID: PMC7409074 DOI: 10.1167/tvst.9.6.20] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose To investigate the intraocular distribution and kinetics of antibodies and nanoparticles in the experimental model. Methods Antibodies (whole IgG 149kDa, antigen-binding fragments 48.39 kDa) and four kinds of nondegradable nanoparticles (25, 50, 200, and 250 nm) were intravitreally injected in the right eye of New Zealand white rabbits. The average optical density and concentration were used to measure intraocular distribution and kinetics. Results After intravitreal injection, antibodies were distributed throughout the vitreous humor and eliminated gradually into anterior and posterior routes. Fluorescence intensity decreased 1 day after injection and was not detected 25 days after injection. The nondegradable nanoparticles migrated posteriorly to the retina 7 days after injection onward and anteriorly to the aqueous humor from 1 hour to 1 day after injection. The fluorescence intensity of the nanoparticles was relatively stable in the vitreous humor, compared to antibodies. Nanoparticles accumulated on the internal limiting membrane of the retina with no penetration into deeper retinal tissue, whereas the smaller size 25 nm nanoparticles passed across the ciliary body and moved into choroid, retina, and suprachoroidal space. A gradual decrease of nanoparticles by their sizes in the vitreous after 30 days after injection was described as the percentage ratio: 61.1% (25 nm), 69.1% (50 nm), 78.6% (200nm), and 85.3% (250 nm). Conclusions Our study revealed the in vivo intraocular distribution and kinetics of antibodies and nanoparticles with diverse sizes and the result might help to develop newer intraocular drugs and drug delivery systems to treat retinal diseases. Translational Relevance These experimental results can be valuable data for human research.
Collapse
Affiliation(s)
- Hyeong Min Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | | | - Hye Kyoung Hong
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yoonha Hwang
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Pilhan Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eunsol Yang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Bundang Hospital, Seongnam, Republic of Korea
| | - Jae Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Bundang Hospital, Seongnam, Republic of Korea
| | - Sunyoung Park
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, Republic of Korea
| | - Young Joo Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kyu Hyung Park
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, Republic of Korea
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
28
|
Zhang H, Rombouts K, Raes L, Xiong R, De Smedt SC, Braeckmans K, Remaut K. Fluorescence-Based Quantification of Messenger RNA and Plasmid DNA Decay Kinetics in Extracellular Biological Fluids and Cell Extracts. ACTA ACUST UNITED AC 2020; 4:e2000057. [PMID: 32402121 DOI: 10.1002/adbi.202000057] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022]
Abstract
Extracellular and intracellular degradation of nucleic acids remains an issue in non-viral gene therapy. Understanding biodegradation is critical for the rational design of gene therapeutics in order to maintain stability and functionality at the target site. However, there are only limited methods available that allow determining the stability of genetic materials in biological environments. In this context, the decay kinetics of fluorescently labeled plasmid DNA (pDNA) and messenger RNA (mRNA) in undiluted biological samples (i.e., human serum, human ascites, bovine vitreous) and cell extracts is studied using fluorescence correlation spectroscopy (FCS) and single particle tracking (SPT). It is demonstrated that FCS is suitable to follow mRNA degradation, while SPT is better suited to investigate pDNA integrity. The half-life of mRNA and pDNA is ≈1-2 min and 1-4 h in biological samples, respectively. The resistance against biodegradation drastically improves by complexation with lipid-based carriers. Taken together, FCS and SPT are able to quantify the integrity of mRNA and pDNA, respectively, as a function of time, both in the extracellular biological fluids and cell extracts. This in turn allows to focus on the important but less understood issue of nucleic acids degradation in more detail and to rationally optimize gene delivery system as therapeutics.
Collapse
Affiliation(s)
- Heyang Zhang
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Koen Rombouts
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Laurens Raes
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| | - Katrien Remaut
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, 9000, Belgium
| |
Collapse
|
29
|
Nayak K, Misra M. PEGylated microemulsion for dexamethasone delivery to posterior segment of eye. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:1071-1090. [PMID: 32149562 DOI: 10.1080/09205063.2020.1740964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Dexamethasone (Dex) is one of the most commonly used anti-vascular endothelial growth factor (anti-VEGF) drugs being used in ocular diseases whether it is associated with anterior segment or posterior segment. For diseases of posterior segment of eye, Dex is delivered as intravitreal implant but the route used for the same is very invasive and poses several hazards on long term use. Thus, topical formulation with ability to outreach retina from ocular surface was intended. Thus, polyethylene glycolylated (PEGylated) microemulsion (ME) was attempted as it can cross the membranous barrier of eye (cornea, conjunctiva, and sclera) and remain afloat in fluidic barrier (aqueous humor, choroid, etc.) as well. Present investigation involved development of Dex-loaded PEGylated ME which was stable, non-toxic to ocular surface, capable to cross cornea and enhanced residence as well as availability of loaded drug in retina. The developed PEGylated ME had physicochemical properties like size (15.98 ± 3.05 nm), polydispersity index (0.25 ± 0.04), zeta potential (-0.04 ± 0.47 mV), percentage transmittance (99.84 ± 1.17%), and drug content (99.32 ± 3.21%). It showed sustained Dex release in in vitro conditions. It also displayed efficiency in enhancing retention of drugs in retina in in vivo pharmacokinetic study on Sprague-Dawley rats. PEGylated ME can retain the drug in retina of rats longer than simple eye drop solution via topical ocular route.
Collapse
Affiliation(s)
- Kritika Nayak
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Gandhinagar, Gujarat, India
| | - Manju Misra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Gandhinagar, Gujarat, India.,Scientist B, B V Patel PERD Centre, Ahmedabad, Gujarat, India
| |
Collapse
|
30
|
Thakur SS, Shenoy SK, Suk JS, Hanes JS, Rupenthal ID. Validation of hyaluronic acid-agar-based hydrogels as vitreous humor mimetics for in vitro drug and particle migration evaluations. Eur J Pharm Biopharm 2020; 148:118-125. [PMID: 31981693 DOI: 10.1016/j.ejpb.2020.01.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/16/2020] [Accepted: 01/19/2020] [Indexed: 12/26/2022]
Abstract
Artificial vitreous humor holds immense potential for use in in vitro intravitreal drug delivery assays. In this study, we investigated rheological properties and drug or nanoparticle migration in hyaluronic acid (HA) - agar based hydrogels and compared these characteristics with bovine vitreous humor. Gel compositions identified in literature containing HA (0.7-5.0 mg/ml) and agar (0.95-4.0 mg/ml) were classified as either high (VH), medium (VM) or low (VL) polymer load. Viscoelastic behavior was evaluated using oscillatory rheology, and migration of differently sized and charged polystyrene nanoparticles (NPs) through the different gels was determined via multiple particle tracking. Comparable rheological behaviour was observed between VL and bovine vitreous. Tracking evaluations revealed that increasing particle size and gel viscosity slowed NP migration. Additionally, 100 nm anionic NPs migrated slower than neutral NPs in VL and VM, while cationic NPs were immobile in all gels. Finally, distribution and clearance of sodium fluorescein was used to model drug mobility through the gels using a custom-built eye model. Flow and angular movement only influenced drug migration in VL and VM, but not VH. Finally, VL and VM demonstrated to have the most similar sodium fluorescein clearance to that of bovine vitreous humor. Together, these evaluations demonstrate that low viscosity HA-agar gels can be used to approximate nanoparticle and drug migration through biological vitreous humor.
Collapse
Affiliation(s)
- Sachin S Thakur
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Siddharth K Shenoy
- Center for Nanomedicine, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - Jung Soo Suk
- Center for Nanomedicine, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - Justin S Hanes
- Center for Nanomedicine, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
31
|
Dubashynskaya N, Poshina D, Raik S, Urtti A, Skorik YA. Polysaccharides in Ocular Drug Delivery. Pharmaceutics 2019; 12:E22. [PMID: 31878298 PMCID: PMC7023054 DOI: 10.3390/pharmaceutics12010022] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/19/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023] Open
Abstract
Polysaccharides, such as cellulose, hyaluronic acid, alginic acid, and chitosan, as well as polysaccharide derivatives, have been successfully used to augment drug delivery in the treatment of ocular pathologies. The properties of polysaccharides can be extensively modified to optimize ocular drug formulations and to obtain biocompatible and biodegradable drugs with improved bioavailability and tailored pharmacological effects. This review discusses the available polysaccharide choices for overcoming the difficulties associated with ocular drug delivery, and it explores the reasons for the dependence between the physicochemical properties of polysaccharide-based drug carriers and their efficiency in different formulations and applications. Polysaccharides will continue to be of great interest to researchers endeavoring to develop ophthalmic drugs with improved effectiveness and safety.
Collapse
Affiliation(s)
- Natallia Dubashynskaya
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St. Petersburg, Russia; (N.D.); (D.P.); (S.R.)
| | - Daria Poshina
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St. Petersburg, Russia; (N.D.); (D.P.); (S.R.)
| | - Sergei Raik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St. Petersburg, Russia; (N.D.); (D.P.); (S.R.)
- Institute of Chemistry, St. Petersburg State University, Universitetskii pr. 26, Petrodvorets, 198504 St. Petersburg, Russia;
| | - Arto Urtti
- Institute of Chemistry, St. Petersburg State University, Universitetskii pr. 26, Petrodvorets, 198504 St. Petersburg, Russia;
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Yury A. Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St. Petersburg, Russia; (N.D.); (D.P.); (S.R.)
- Institute of Chemistry, St. Petersburg State University, Universitetskii pr. 26, Petrodvorets, 198504 St. Petersburg, Russia;
| |
Collapse
|
32
|
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: 5.3] [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
|
33
|
Esteban-Pérez S, Bravo-Osuna I, Andrés-Guerrero V, Molina-Martínez IT, Herrero-Vanrell R. Trojan Microparticles Potential for Ophthalmic Drug Delivery. Curr Med Chem 2019; 27:570-582. [PMID: 31486746 DOI: 10.2174/0929867326666190905150331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/11/2019] [Accepted: 08/30/2019] [Indexed: 11/22/2022]
Abstract
The administration of drugs to treat ocular disorders still remains a technological challenge in this XXI century. Although there is an important arsenal of active molecules useful to treat ocular diseases, ranging from classical compounds to biotechnological products, currenty, no ideal delivery system is able to profit all their therapeutic potential. Among the Intraocular Drug Delivery Systems (IODDS) proposed to overcome some of the most important limitations, microsystems and nanosystems have raised high attention. While microsystems are able to offer long-term release after intravitreal injection, nanosystems can protect the active compound from external environment (reducing their clearance) and direct it to its target tissues. In recent years, some researchers have explored the possibility of combining micro and nanosystems in "Nanoparticle-in-Microparticle (NiMs)" systems or "trojan systems". This excellent idea is not exempt of technological problems, remains partially unsolved, especially in the case of IODDS. The objective of the present review is to show the state of art concerning the design, preparation and characterization of trojan microparticles for drug delivery and to remark their potential and limitations as IODDS, one of the most important challenges faced by pharmaceutical technology at the moment.
Collapse
Affiliation(s)
- Sergio Esteban-Pérez
- Complutense University, InnOftal Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramon y Cajal s/n, Madrid 28040, Spain
| | - Irene Bravo-Osuna
- Complutense University, InnOftal Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramon y Cajal s/n, Madrid 28040, Spain.,Ocular Pathology National Net (OFTARED) of the Institute of Health Carlos III, Health Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Vanessa Andrés-Guerrero
- Complutense University, InnOftal Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramon y Cajal s/n, Madrid 28040, Spain
| | - Irene T Molina-Martínez
- Complutense University, InnOftal Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramon y Cajal s/n, Madrid 28040, Spain
| | - Rocío Herrero-Vanrell
- Complutense University, InnOftal Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramon y Cajal s/n, Madrid 28040, Spain
| |
Collapse
|
34
|
Pai RV, Monpara JD, Vavia PR. Exploring molecular dynamics simulation to predict binding with ocular mucin: An in silico approach for screening mucoadhesive materials for ocular retentive delivery systems. J Control Release 2019; 309:190-202. [DOI: 10.1016/j.jconrel.2019.07.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 01/13/2023]
|
35
|
Subrizi A, del Amo EM, Korzhikov-Vlakh V, Tennikova T, Ruponen M, Urtti A. Design principles of ocular drug delivery systems: importance of drug payload, release rate, and material properties. Drug Discov Today 2019; 24:1446-1457. [DOI: 10.1016/j.drudis.2019.02.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/10/2019] [Accepted: 02/01/2019] [Indexed: 12/26/2022]
|
36
|
Sauvage F, Fraire JC, Remaut K, Sebag J, Peynshaert K, Harrington M, Van de Velde FJ, Xiong R, Tassignon MJ, Brans T, Braeckmans K, De Smedt SC. Photoablation of Human Vitreous Opacities by Light-Induced Vapor Nanobubbles. ACS NANO 2019; 13:8401-8416. [PMID: 31287662 DOI: 10.1021/acsnano.9b04050] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Myopia, diabetes, and aging are the main causes of progressive vitreous collagen aggregation, resulting in vitreous opacities, which can significantly disturb vision. As vitreous opacities, which induce the visual phenomenon of "floaters", are accessible with nanomaterials and light, we propose a nanotechnology-based approach to locally ablate them with highly reduced light energy compared to the more traditional YAG laser therapy. Our strategy relies on the plasmon properties of gold nanoparticles that generate vapor nanobubbles upon pulsed-laser illumination whose mechanical force can ablate vitreous opacities. We designed gold nanoparticles coated with hyaluronic acid (HA), which have excellent diffusional mobility in human vitreous, an essential requirement to reach the vitreous opacities. In addition, we found that HA-coated gold nanoparticles can accumulate extensively on human vitreous opacities that were obtained by vitrectomy from patients with vision-degrading myodesopsia. When subsequently applying nanosecond laser pulses, the collagen aggregates were efficiently destroyed with ∼1000 times less light energy than typically used in YAG laser therapy. This low-energy "floater-specific destruction", which is due to the accumulation of the small gold nanoparticles on the opacities, is attractive, as it may be safer to the surrounding ocular tissues while at the same time being easier and faster to apply compared to YAG laser therapy, where the opacities need to be ablated piece by piece by a tightly focused laser beam. Gold nanoparticle-assisted photoablation may therefore provide a safer, faster, and more reliable destruction of vitreous opacities in the treatment of ophthalmologic diseases.
Collapse
Affiliation(s)
- Félix Sauvage
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| | - Juan C Fraire
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| | - Katrien Remaut
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| | - J Sebag
- VMR Institute for Vitreous Macula Retina , Huntington Beach , California 92647 , United States
- Doheny Eye Institute/UCLA , Los Angeles , California 90033 , United States
| | - Karen Peynshaert
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| | - Michael Harrington
- Huntington Medical Research Institutes , Pasadena , California 91105 , United States
| | - Frans J Van de Velde
- Schepens Eye Research Institute , Harvard Medical School , Boston , Massachusetts 02114 , United States
| | - Ranhua Xiong
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| | - Marie-José Tassignon
- Department of Ophthalmology, Antwerp University Hospital , University of Antwerp , Antwerp 2020 , Belgium
| | - Toon Brans
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry & Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent 9000 , Belgium
| |
Collapse
|
37
|
Devoldere J, Peynshaert K, Dewitte H, Vanhove C, De Groef L, Moons L, Özcan SY, Dalkara D, De Smedt SC, Remaut K. Non-viral delivery of chemically modified mRNA to the retina: Subretinal versus intravitreal administration. J Control Release 2019; 307:315-330. [PMID: 31265881 DOI: 10.1016/j.jconrel.2019.06.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/20/2019] [Accepted: 06/29/2019] [Indexed: 12/20/2022]
Abstract
mRNA therapeutics have recently experienced a new wave of interest, mainly due to the discovery that chemical modifications to mRNA's molecular structure could drastically reduce its inherent immunogenicity and perceived instability. On this basis, we aimed to explore the potential of chemically stabilized mRNA for ocular applications. More specifically, we investigated the behavior of mRNA-loaded lipid-based carriers in human retinal cells (in vitro), in bovine retinal explants (ex vivo) and in mouse retinas (in vivo). We demonstrate a clear superiority of mRNA over pDNA to induce protein expression in different retinal cell types, which was further enhanced by chemical modification of the mRNA, providing up to ~1800-fold higher reporter gene expression compared to pDNA. Moreover, transgene expression could be detected for at least 20 days after a single administration of chemically modified mRNA in vitro. We furthermore determined the localization and extent of mRNA expression depending on the administration route. After subretinal (SR) administration, mRNA expression was observed in vivo and ex vivo. By contrast, intravitreal (IVT) administration resulted in limited expression in vivo. Using ex vivo bovine explants with an intact vitreoretinal (VR) interface we could attribute this to the inner limiting membrane (ILM), which presents a large barrier for non-viral delivery of mRNA, trapping mRNA complexes at the vitreal side. When the vitreous was removed, which compromises the ILM, mRNA expression was apparent and seemed to colocalize with Müller cells or photoreceptors after respectively IVT or SR administration. Taken together, this study represents a first step towards mRNA-mediated therapy for retinal diseases.
Collapse
Affiliation(s)
- Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Medical School of the Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1050 Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Christian Vanhove
- Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Sinem Yilmaz Özcan
- Neurological Sciences and Psychiatry Institute; Hacettepe University, Ankara, Turkey
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
38
|
Devoldere J, Wels M, Peynshaert K, Dewitte H, De Smedt SC, Remaut K. The obstacle course to the inner retina: Hyaluronic acid-coated lipoplexes cross the vitreous but fail to overcome the inner limiting membrane. Eur J Pharm Biopharm 2019; 141:161-171. [PMID: 31150809 DOI: 10.1016/j.ejpb.2019.05.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/27/2019] [Accepted: 05/27/2019] [Indexed: 10/26/2022]
Abstract
Considerable research over the last few years has revealed dysregulation of growth factors in various retinal diseases, such as glaucoma, diabetic retinopathy and photoreceptor degenerations. The use of messengerRNA (mRNA) to transiently overexpress a specific factor could compensate for this imbalance. However, a critical challenge of this approach lies in the ability to efficiently deliver mRNA molecules to the retinal target cells. In this study we found that intravitreal (IVT) injection is an attractive approach to deliver mRNA to the retina, providing two critical barriers can be overcome: the vitreous and the inner limiting membrane (ILM). We demonstrated that the vitreous is indeed a major hurdle in the delivery of the cationic mRNA-complexes to retinal cells, both in terms of vitreal mobility and cellular uptake. To improve their intravitreal mobility and avoid unwanted extracellular interactions, we evaluated the use of hyaluronic acid (HA) as an electrostatic coating strategy. This HA-coating provided the complexes with a negative surface charge, markedly enhancing their mobility in the vitreous humor, without reducing their cellular internalization and transfection efficiency. However, although this coating strategy allows the mRNA-complexes to successfully overcome the vitreal barrier, the majority of the particles accumulated at the ILM. This study therefore underscores the crucial barrier function of the ILM toward non-viral retinal gene delivery and the need to smartly design mRNA-carriers able to surmount the vitreous as well as the ILM.
Collapse
Affiliation(s)
- Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Mike Wels
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Medical School of the Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1050 Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
39
|
Subia B, Reinisalo M, Dey N, Tavakoli S, Subrizi A, Ganguli M, Ruponen M. Nucleic acid delivery to differentiated retinal pigment epithelial cells using cell-penetrating peptide as a carrier. Eur J Pharm Biopharm 2019; 140:91-99. [PMID: 31085311 DOI: 10.1016/j.ejpb.2019.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/16/2019] [Accepted: 05/08/2019] [Indexed: 10/26/2022]
Abstract
Nucleic acid delivery to the eye is a promising treatment strategy for many retinal disorders. In this manuscript, retinal gene delivery with non-coated and chondroitin sulphate (CS) coated amphipathic and cationic peptides was tested. The transfection and gene knockdown efficiencies were evaluated in different retinal pigment epithelial (RPE) cell models including both dividing and differentiated cells. In addition, the mobility of peptide-based gene delivery systems was examined in porcine vitreous by particle tracking analysis. The results indicate that amphipathic and cationic peptides are safe in vitro and are capable of high transgene expression and gene knockdown in dividing cells. We further demonstrate that incorporation of CS improves the efficiency of gene delivery of peptide-based systems. Most importantly, the transgene expression mediated by both non-coated and CS coated peptides was high in differentiated as well as in human primary RPE cells which are typically difficult to transfect. Coating of peptide-based gene delivery systems with CS improved diffusion in the vitreous and enhanced the stability of the polyplexes. The results indicate that a peptide-based system can be fine-tuned as a promising approach for retinal gene delivery.
Collapse
Affiliation(s)
- Bano Subia
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland.
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland
| | - Namit Dey
- Delhi Technological University, Delhi 110042, India
| | | | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C 800, Denmark
| | - Munia Ganguli
- CSIR-Institute of Genomics and Integrative Biology, New Delhi 110021, India
| | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland
| |
Collapse
|
40
|
Light-Triggered Cellular Delivery of Oligonucleotides. Pharmaceutics 2019; 11:pharmaceutics11020090. [PMID: 30795565 PMCID: PMC6410276 DOI: 10.3390/pharmaceutics11020090] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/01/2019] [Accepted: 02/14/2019] [Indexed: 01/09/2023] Open
Abstract
The major challenge in the therapeutic applicability of oligonucleotide-based drugs is the development of efficient and safe delivery systems. The carriers should be non-toxic and stable in vivo, but interact with the target cells and release the loaded oligonucleotides intracellularly. We approached this challenge by developing a light-triggered liposomal delivery system for oligonucleotides based on a non-cationic and thermosensitive liposome with indocyanine green (ICG) as photosensitizer. The liposomes had efficient release properties, as 90% of the encapsulated oligonucleotides were released after 1-minute light exposure. Cell studies using an enhanced green fluorescent protein (EGFP)-based splicing assay with HeLa cells showed light-activated transfection with up to 70%–80% efficacy. Moreover, free ICG and oligonucleotides in solution transfected cells upon light induction with similar efficacy as the liposomal system. The light-triggered delivery induced moderate cytotoxicity (25%–35% reduction in cell viability) 1–2 days after transfection, but the cell growth returned to control levels in 4 days. In conclusion, the ICG-based light-triggered delivery is a promising method for oligonucleotides, and it can be used as a platform for further optimization and development.
Collapse
|
41
|
Bohórquez AC, Unni M, Belsare S, Chiu-Lam A, Rice L, Pampo C, Siemann D, Rinaldi C. Stability and Mobility of Magnetic Nanoparticles in Biological Environments Determined from Dynamic Magnetic Susceptibility Measurements. Bioconjug Chem 2018; 29:2793-2805. [PMID: 30011185 DOI: 10.1021/acs.bioconjchem.8b00419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Low tumor accumulation following systemic delivery remains a key challenge for advancing many cancer nanomedicines. One obstacle in engineering nanoparticles for high tumor accumulation is a lack of techniques to monitor their stability and mobility in situ. One way to monitor the stability and mobility of magnetic nanoparticles biological fluids in situ is through dynamic magnetic susceptibility measurements (DMS), which under certain conditions provide a measure of the particle's rotational diffusivity. For magnetic nanoparticles modified to have commonly used biomedical surface coatings, we describe a systematic comparison of DMS measurements in whole blood and tumor tissue explants. DMS measurements clearly demonstrated that stability and mobility changed over time and from one medium to another for each different coating. It was found that nanoparticles coated with covalently grafted, dense layers of PEG were the only ones to show good stability and mobility in all settings tested. These studies illustrate the utility of DMS measurements to estimate the stability and mobility of nanoparticles in situ, and which can provide insights that lead to engineering better nanoparticles for in vivo use.
Collapse
Affiliation(s)
- Ana C Bohórquez
- J. Crayton Pruitt Family Department of Biomedical Engineering , University of Florida , 1275 Center Drive , Biomedical Sciences Building, Gainesville , Florida 32611 , United States
| | - Mythreyi Unni
- Department of Chemical Engineering , University of Florida , 1030 Center Drive , Gainesville , Florida 32611 , United States
| | - Sayali Belsare
- J. Crayton Pruitt Family Department of Biomedical Engineering , University of Florida , 1275 Center Drive , Biomedical Sciences Building, Gainesville , Florida 32611 , United States
| | - Andreina Chiu-Lam
- Department of Chemical Engineering , University of Florida , 1030 Center Drive , Gainesville , Florida 32611 , United States
| | - Lori Rice
- Department of Radiation Oncology , University of Florida , Gainesville , Florida 32610 , United States
| | - Christine Pampo
- Department of Radiation Oncology , University of Florida , Gainesville , Florida 32610 , United States
| | - Dietmar Siemann
- Department of Radiation Oncology , University of Florida , Gainesville , Florida 32610 , United States
| | - Carlos Rinaldi
- J. Crayton Pruitt Family Department of Biomedical Engineering , University of Florida , 1275 Center Drive , Biomedical Sciences Building, Gainesville , Florida 32611 , United States.,Department of Chemical Engineering , University of Florida , 1030 Center Drive , Gainesville , Florida 32611 , United States
| |
Collapse
|
42
|
Huang D, Chen YS, Xu Q, Hanes J, Rupenthal ID. Effects of enzymatic degradation on dynamic mechanical properties of the vitreous and intravitreal nanoparticle mobility. Eur J Pharm Sci 2018; 118:124-133. [DOI: 10.1016/j.ejps.2018.03.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 11/15/2022]
|
43
|
Huang D, Chen YS, Green CR, Rupenthal ID. Hyaluronic acid coated albumin nanoparticles for targeted peptide delivery in the treatment of retinal ischaemia. Biomaterials 2018; 168:10-23. [DOI: 10.1016/j.biomaterials.2018.03.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 12/19/2022]
|
44
|
Peynshaert K, Devoldere J, De Smedt SC, Remaut K. In vitro and ex vivo models to study drug delivery barriers in the posterior segment of the eye. Adv Drug Deliv Rev 2018; 126:44-57. [PMID: 28939376 DOI: 10.1016/j.addr.2017.09.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/18/2017] [Accepted: 09/08/2017] [Indexed: 12/18/2022]
Abstract
Many ocular disorders leading to blindness could benefit from efficient delivery of therapeutics to the retina. However, despite extensive research into drug delivery vehicles and administration techniques, efficacy remains limited because of the many static and dynamic barriers present in the eye. Comprehension of the various barriers and especially how to overcome them can improve our ability to estimate the potential of existent drug delivery vectors and support the design of new ones. To this end, this review gives an overview of the most important ocular barriers for each administration route to the back of the eye. For each barrier, its biological composition and its role as an obstacle towards macromolecules, nanoparticles and viral vectors will be discussed; special attention will be paid to the influence of size, charge and lipophilicity of drug(s) (carrier) on their ability to overcome each barrier. Finally, the most significant available in vitro and ex vivo methods and models to test the potential of a therapeutic to cross each barrier are listed.
Collapse
|
45
|
Peynshaert K, Devoldere J, Forster V, Picaud S, Vanhove C, De Smedt SC, Remaut K. Toward smart design of retinal drug carriers: a novel bovine retinal explant model to study the barrier role of the vitreoretinal interface. Drug Deliv 2017; 24:1384-1394. [PMID: 28925755 PMCID: PMC8241179 DOI: 10.1080/10717544.2017.1375578] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022] Open
Abstract
Retinal gene delivery via intravitreal injection is hampered by various physiological barriers present in the eye of which the vitreoretinal (VR) interface represents the most serious hurdle. In this study, we present a retinal explant model especially designed to study the role of this interface as a barrier for the penetration of vectors into the retina. In contrast to all existing explant models, the developed model is bovine-derived and more importantly, keeps the vitreous attached to the retina at all times to guarantee an intact VR interface. After ex vivo intravitreal injection into the living retinal explant, the route of fluorescent carriers across the VR interface can be tracked. By applying two different imaging methods on this model, we discovered that the transfer through the VR barrier is size-dependent since 40 nm polystyrene particles are more easily taken up in the retina than 100 and 200 nm sized particles. In addition, we found that removing the vitreous, as commonly done for culture of conventional explants, leads to an overestimation of particle uptake, and conclude that the ultimate barrier to overcome for retinal uptake is undoubtedly the inner limiting membrane. Damaging this matrix resulted in a massive increase in particle transfer into the retina. In conclusion, we have developed a highly relevant ex vivo model that maximally mimics the human in vivo physiology which can be applied as a representative test set-up to assess the potential of promising drug delivery carriers to cross the VR interface.
Collapse
Affiliation(s)
- Karen Peynshaert
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
| | - Joke Devoldere
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
| | - Valérie Forster
- Institut de la Vision, INSERM, Université Paris 6, Paris, France
| | - Serge Picaud
- Institut de la Vision, INSERM, Université Paris 6, Paris, France
| | - Christian Vanhove
- Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Stefaan C. De Smedt
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
| | - Katrien Remaut
- Lab of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
| |
Collapse
|
46
|
Huang D, Chen YS, Thakur SS, Rupenthal ID. Ultrasound-mediated nanoparticle delivery across ex vivo bovine retina after intravitreal injection. Eur J Pharm Biopharm 2017; 119:125-136. [DOI: 10.1016/j.ejpb.2017.06.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/06/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022]
|
47
|
Witten J, Ribbeck K. The particle in the spider's web: transport through biological hydrogels. NANOSCALE 2017; 9:8080-8095. [PMID: 28580973 PMCID: PMC5841163 DOI: 10.1039/c6nr09736g] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Biological hydrogels such as mucus, extracellular matrix, biofilms, and the nuclear pore have diverse functions and compositions, but all act as selectively permeable barriers to the diffusion of particles. Each barrier has a crosslinked polymeric mesh that blocks penetration of large particles such as pathogens, nanotherapeutics, or macromolecules. These polymeric meshes also employ interactive filtering, in which affinity between solutes and the gel matrix controls permeability. Interactive filtering affects the transport of particles of all sizes including peptides, antibiotics, and nanoparticles and in many cases this filtering can be described in terms of the effects of charge and hydrophobicity. The concepts described in this review can guide strategies to exploit or overcome gel barriers, particularly for applications in diagnostics, pharmacology, biomaterials, and drug delivery.
Collapse
Affiliation(s)
- Jacob Witten
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | |
Collapse
|
48
|
Santos RS, Dakwar GR, Zagato E, Brans T, Figueiredo C, Raemdonck K, Azevedo NF, De Smedt SC, Braeckmans K. Intracellular delivery of oligonucleotides in Helicobacter pylori by fusogenic liposomes in the presence of gastric mucus. Biomaterials 2017; 138:1-12. [PMID: 28550752 DOI: 10.1016/j.biomaterials.2017.05.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
Abstract
The rising antimicrobial resistance contributes to 25000 annual deaths in Europe. This threat to the public health can only be tackled if novel antimicrobials are developed, combined with a more precise use of the currently available antibiotics through the implementation of fast, specific, diagnostic methods. Nucleic acid mimics (NAMs) that are able to hybridize intracellular bacterial RNA have the potential to become such a new class of antimicrobials and additionally could serve as specific detection probes. However, an essential requirement is that these NAMs should be delivered into the bacterial cytoplasm, which is a particular challenge given the fact that they are charged macromolecules. We consider these delivery challenges in relation to the gastric pathogen Helicobacter pylori, the most frequent chronic infection worldwide. In particular, we evaluate if cationic fusogenic liposomes are suitable carriers to deliver NAMs across the gastric mucus barrier and the bacterial envelope. Our study shows that DOTAP-DOPE liposomes post-PEGylated with DSPE-PEG (DSPE Lpx) can indeed successfully deliver NAMs into Helicobacter pylori, while offering protection to the NAMs from binding and inactivation in gastric mucus isolated from pigs. DSPE Lpx thus offer exciting new possibilities for in vivo diagnosis and treatment of Helicobacter pylori infections.
Collapse
MESH Headings
- Animals
- Anti-Infective Agents/administration & dosage
- Anti-Infective Agents/chemical synthesis
- Anti-Infective Agents/metabolism
- Cytoplasm/metabolism
- Drug Delivery Systems
- Drug Resistance, Microbial
- Fatty Acids, Monounsaturated/chemistry
- Fluorescent Dyes/chemistry
- Helicobacter Infections/diagnosis
- Helicobacter Infections/drug therapy
- Helicobacter Infections/microbiology
- Helicobacter pylori/genetics
- Helicobacter pylori/metabolism
- In Situ Hybridization, Fluorescence
- Liposomes
- Molecular Mimicry
- Mucus/chemistry
- Mucus/microbiology
- Oligonucleotides/administration & dosage
- Oligonucleotides/chemical synthesis
- Oligonucleotides/genetics
- Oligonucleotides/metabolism
- Oligonucleotides, Antisense/administration & dosage
- Oligonucleotides, Antisense/chemical synthesis
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/metabolism
- Phosphatidylethanolamines/chemistry
- Polyethylene Glycols/chemistry
- Quaternary Ammonium Compounds/chemistry
- RNA, Bacterial/antagonists & inhibitors
- RNA, Bacterial/genetics
- RNA, Ribosomal/antagonists & inhibitors
- RNA, Ribosomal/genetics
- Stomach/microbiology
- Swine
Collapse
Affiliation(s)
- Rita S Santos
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium; LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, Porto, Portugal; i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - George R Dakwar
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Elisa Zagato
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium; Center for Nano- and Biophotonics, Ghent University, Ghent, Belgium
| | - Toon Brans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium; Center for Nano- and Biophotonics, Ghent University, Ghent, Belgium
| | - Céu Figueiredo
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal; Department of Pathology and Oncology, Faculty of Medicine of the University of Porto, Portugal
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Nuno F Azevedo
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, Porto, Portugal
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium.
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium; Center for Nano- and Biophotonics, Ghent University, Ghent, Belgium
| |
Collapse
|
49
|
Mixtures of hyaluronic acid and liposomes for drug delivery: Phase behavior, microstructure and mobility of liposomes. Int J Pharm 2017; 523:246-259. [DOI: 10.1016/j.ijpharm.2017.03.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/03/2017] [Accepted: 03/16/2017] [Indexed: 01/25/2023]
|
50
|
Martens TF, Peynshaert K, Nascimento TL, Fattal E, Karlstetter M, Langmann T, Picaud S, Demeester J, De Smedt SC, Remaut K, Braeckmans K. Effect of hyaluronic acid-binding to lipoplexes on intravitreal drug delivery for retinal gene therapy. Eur J Pharm Sci 2017; 103:27-35. [DOI: 10.1016/j.ejps.2017.02.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 11/17/2022]
|