1
|
Adamska JM, Leftheriotis S, Bosma R, Vischer HF, Leurs R. Multiplex Detection of Fluorescent Chemokine Binding to CXC Chemokine Receptors by NanoBRET. Int J Mol Sci 2024; 25:5018. [PMID: 38732237 PMCID: PMC11084278 DOI: 10.3390/ijms25095018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
NanoLuc-mediated bioluminescence resonance energy transfer (NanoBRET) has gained popularity for its ability to homogenously measure ligand binding to G protein-coupled receptors (GPCRs), including the subfamily of chemokine receptors. These receptors, such as ACKR3, CXCR4, CXCR3, play a crucial role in the regulation of the immune system, are associated with inflammatory diseases and cancer, and are seen as promising drug targets. The aim of this study was to optimize NanoBRET-based ligand binding to NLuc-ACKR3 and NLuc-CXCR4 using different fluorescently labeled chemokine CXCL12 analogs and their use in a multiplex NanoBRET binding assay of two chemokine receptors at the same time. The four fluorescent CXCL12 analogs (CXCL12-AZD488, -AZD546, -AZD594, -AZD647) showed high-affinity saturable binding to both NLuc-ACKR3 and NLuc-CXCR4, with relatively low levels of non-specific binding. Additionally, the binding of all AZDye-labeled CXCL12s to Nluc receptors was inhibited by pharmacologically relevant unlabeled chemokines and small molecules. The NanoBRET binding assay for CXCL10-AZD488 binding to Nluc-CXCR3 was also successfully established and successfully employed for the simultaneous measurement of the binding of unlabeled small molecules to NLuc-CXCR3 and NLuc-CXCR4. In conclusion, multiplexing the NanoBRET-based competition binding assay is a promising tool for testing unlabeled (small) molecules against multiple GPCRs simultaneously.
Collapse
Affiliation(s)
| | | | | | | | - Rob Leurs
- Amsterdam Institute of Molecular and Life Sciences, Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands (S.L.); (H.F.V.)
| |
Collapse
|
2
|
Shastri DH, Silva AC, Almeida H. Ocular Delivery of Therapeutic Proteins: A Review. Pharmaceutics 2023; 15:pharmaceutics15010205. [PMID: 36678834 PMCID: PMC9864358 DOI: 10.3390/pharmaceutics15010205] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/25/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Therapeutic proteins, including monoclonal antibodies, single chain variable fragment (ScFv), crystallizable fragment (Fc), and fragment antigen binding (Fab), have accounted for one-third of all drugs on the world market. In particular, these medicines have been widely used in ocular therapies in the treatment of various diseases, such as age-related macular degeneration, corneal neovascularization, diabetic retinopathy, and retinal vein occlusion. However, the formulation of these biomacromolecules is challenging due to their high molecular weight, complex structure, instability, short half-life, enzymatic degradation, and immunogenicity, which leads to the failure of therapies. Various efforts have been made to overcome the ocular barriers, providing effective delivery of therapeutic proteins, such as altering the protein structure or including it in new delivery systems. These strategies are not only cost-effective and beneficial to patients but have also been shown to allow for fewer drug side effects. In this review, we discuss several factors that affect the design of formulations and the delivery of therapeutic proteins to ocular tissues, such as the use of injectable micro/nanocarriers, hydrogels, implants, iontophoresis, cell-based therapy, and combination techniques. In addition, other approaches are briefly discussed, related to the structural modification of these proteins, improving their bioavailability in the posterior segments of the eye without affecting their stability. Future research should be conducted toward the development of more effective, stable, noninvasive, and cost-effective formulations for the ocular delivery of therapeutic proteins. In addition, more insights into preclinical to clinical translation are needed.
Collapse
Affiliation(s)
- Divyesh H. Shastri
- Department of Pharmaceutics & Pharmaceutical Technology, K.B. Institute of Pharmaceutical Education and Research, Kadi Sarva Vishwavidyalaya, Sarva Vidyalaya Kelavani Mandal, Gandhinagar 382016, India
- Correspondence:
| | - Ana Catarina Silva
- FP-I3ID (Instituto de Investigação, Inovação e Desenvolvimento), FP-BHS (Biomedical and Health Sciences Research Unit), Faculty of Health Sciences, University Fernando Pessoa, 4249-004 Porto, Portugal
- UCIBIO (Research Unit on Applied Molecular Biosciences), REQUIMTE (Rede de Química e Tecnologia), MEDTECH (Medicines and Healthcare Products), Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Hugo Almeida
- UCIBIO (Research Unit on Applied Molecular Biosciences), REQUIMTE (Rede de Química e Tecnologia), MEDTECH (Medicines and Healthcare Products), Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Mesosystem Investigação & Investimentos by Spinpark, Barco, 4805-017 Guimarães, Portugal
| |
Collapse
|
3
|
Maurya SK, Khan P, Rehman AU, Kanchan RK, Perumal N, Mahapatra S, Chand HS, Santamaria-Barria JA, Batra SK, Nasser MW. Rethinking the chemokine cascade in brain metastasis: Preventive and therapeutic implications. Semin Cancer Biol 2022; 86:914-930. [PMID: 34968667 PMCID: PMC9234104 DOI: 10.1016/j.semcancer.2021.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 01/27/2023]
Abstract
Brain metastasis (BrM) is one of the major causes of death in cancer patients and is associated with an estimated 10-40 % of total cancer cases. The survival rate of brain metastatic patients has not improved due to intratumor heterogeneity, the survival adaptations of brain homing metastatic cells, and the lack of understanding of underlying molecular mechanisms that limit the availability of effective therapies. The heterogeneous population of immune cells and tumor-initiating cells or cancer stem cells in the tumor microenvironment (TME) release various factors, such as chemokines that upon binding to their cognate receptors enhance tumor growth at primary sites and help tumor cells metastasize to the brain. Furthermore, brain metastatic sites have unique heterogeneous microenvironment that fuels cancer cells in establishing BrM. This review explores the crosstalk of chemokines with the heterogeneous TME during the progression of BrM and recognizes potential therapeutic approaches. We also discuss and summarize different targeted, immunotherapeutic, chemotherapeutic, and combinatorial strategies (with chemo-/immune- or targeted-therapies) to attenuate chemokines mediated BrM.
Collapse
Affiliation(s)
- Shailendra Kumar Maurya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Parvez Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Asad Ur Rehman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Ranjana K Kanchan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Naveenkumar Perumal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Sidharth Mahapatra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA; Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, 68108, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Hitendra S Chand
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | | | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68108, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA.
| |
Collapse
|
4
|
Sodium butyrate-loaded nanoparticles coated with chitosan for the treatment of neovascularization in age-related macular degeneration: ocular biocompatibility and antiangiogenic activity. Eur J Pharm Biopharm 2022; 179:26-36. [PMID: 36041595 DOI: 10.1016/j.ejpb.2022.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/13/2022] [Accepted: 08/22/2022] [Indexed: 12/29/2022]
Abstract
Sodium butyrate-loaded nanoparticles coated chitosan (NaBu-loaded nanoparticles/CS) were developed to treat the choroidal neovascularization in wet age-related macular degeneration (AMD). The nanoparticles were produced by double emulsification and solvent evaporation technique, optimized by experimental statistical design, characterized by analytical methods, investigated in terms of in vitro and in vivo ocular biocompatibility, and evaluated as an antiangiogenic system in vivo. The NaBu-loaded nanoparticles/CS were 311.1 ± 3.1 nm in diameter with a 0.208 ± 0.007 polydispersity index; had a +56.3 ± 2.6 mV zeta potential; showed a 92.3% NaBu encapsulation efficiency; and sustained the drug release over 35 days. The NaBu-loaded nanoparticles/CS showed no toxicity to human retinal pigment epithelium cells (ARPE-19 cells); was not irritant to the chorioallantoic membrane (CAM); did not interfere in the integrity of the retinal layers of rat's eyes, as detected by the Optical Coherence Tomography and histopathology; and inhibited the angiogenesis in CAM assay. The NaBu-loaded nanoparticles/CS could be a therapeutic alternative to limit the neovascularization in AMD.
Collapse
|
5
|
Das S, Chen Y, Yan J, Christensen G, Belhadj S, Tolone A, Paquet-Durand F. The role of cGMP-signalling and calcium-signalling in photoreceptor cell death: perspectives for therapy development. Pflugers Arch 2021; 473:1411-1421. [PMID: 33864120 PMCID: PMC8370896 DOI: 10.1007/s00424-021-02556-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
The second messengers, cGMP and Ca2+, have both been implicated in retinal degeneration; however, it is still unclear which of the two is most relevant for photoreceptor cell death. This problem is exacerbated by the close connections and crosstalk between cGMP-signalling and calcium (Ca2+)-signalling in photoreceptors. In this review, we summarize key aspects of cGMP-signalling and Ca2+-signalling relevant for hereditary photoreceptor degeneration. The topics covered include cGMP-signalling targets, the role of Ca2+ permeable channels, relation to energy metabolism, calpain-type proteases, and how the related metabolic processes may trigger and execute photoreceptor cell death. A focus is then put on cGMP-dependent mechanisms and how exceedingly high photoreceptor cGMP levels set in motion cascades of Ca2+-dependent and independent processes that eventually bring about photoreceptor cell death. Finally, an outlook is given into mutation-independent therapeutic approaches that exploit specific features of cGMP-signalling. Such approaches might be combined with suitable drug delivery systems for translation into clinical applications.
Collapse
Affiliation(s)
- Soumyaparna Das
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Jie Yan
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Gustav Christensen
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Soumaya Belhadj
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Arianna Tolone
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - François Paquet-Durand
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany.
| |
Collapse
|
6
|
du Toit LC, Choonara YE, Pillay V. An Injectable Nano-Enabled Thermogel to Attain Controlled Delivery of p11 Peptide for the Potential Treatment of Ocular Angiogenic Disorders of the Posterior Segment. Pharmaceutics 2021; 13:pharmaceutics13020176. [PMID: 33525495 PMCID: PMC7910951 DOI: 10.3390/pharmaceutics13020176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 11/16/2022] Open
Abstract
This investigation focused on the design of an injectable nano-enabled thermogel (nano-thermogel) system to attain controlled delivery of p11 anti-angiogenic peptide for proposed effective prevention of neovascularisation and to overcome the drawbacks of the existing treatment approaches for ocular disorders characterised by angiogenesis, which employ multiple intravitreal injections of anti-vascular endothelial growth factor (anti-VEGF) antibodies. Synthesis of a polyethylene glycol-polycaprolactone-polyethylene glycol (PEG-PCL-PEG) triblock co-polymer was undertaken, followed by characterisation employing Fourier-transform infrared (FTIR) spectroscopy, nuclear magnetic resonance (NMR) spectroscopy and differential scanning calorimetry (DSC) to ascertain the chemical stability and integrity of the co-polymer instituted for nano-thermogel formulation. The p11 anti-angiogenic peptide underwent encapsulation within poly(lactic-co-glycolic acid) (PLGA) nanoparticles via a double emulsion solvent evaporation method and was incorporated into the thermogel following characterisation by scanning electron microscopy (SEM), zeta size and zeta-potential analysis. The tube inversion approach and rheological analysis were employed to ascertain the thermo-sensitive sol-gel conversion of the nano-thermogel system. Chromatographic assessment of the in vitro release of the peptide was performed, with stability confirmation via Tris-Tricine PAGE (Polyacrylamide Gel Electrophoresis). In vitro biocompatibility of the nano-thermogel system was investigated employing a retinal cell line (ARP-19). A nanoparticle size range of 100–200 nm and peptide loading efficiency of 67% was achieved. Sol-gel conversion of the nano-thermogel was observed between 32–45 °C. Release of the peptide in vitro was sustained, with maintenance of stability, for 60 days. Biocompatibility assessment highlighted 97–99% cell viability with non-haemolytic ability, which supports the potential applicability of the nano-thermogel system for extended delivery of peptide for ocular disorder treatment.
Collapse
Affiliation(s)
| | - Yahya Essop Choonara
- Correspondence: ; Tel.: +27-11-717-2052; Fax: +27-11-642-4355 or +27-86-553-4733
| | | |
Collapse
|
7
|
DeJulius C, Bernardo-Colón A, Naguib S, Backstrom J, Kavanaugh T, Gupta M, Duvall C, Rex T. Microsphere antioxidant and sustained erythropoietin-R76E release functions cooperate to reduce traumatic optic neuropathy. J Control Release 2021; 329:762-773. [PMID: 33049330 PMCID: PMC8162757 DOI: 10.1016/j.jconrel.2020.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 01/12/2023]
Abstract
Wild-type erythropoietin (EPO) is promising for neuroprotection, but its therapeutic use is limited because it causes a systemic rise in hematocrit. We have developed an EPO-R76E derivative that maintains neuroprotective function without effects on hematocrit, but this protein has a short half-life in vivo. Here, we compare the efficacy and carrier-induced inflammatory response of two polymeric microparticle (MP) EPO-R76E sustained release formulations based on conventional hydrolytically degradable poly(lactic-co-glycolic acid) (PLGA) and reactive oxygen species (ROS)-degradable poly(propylene sulfide) (PPS). Both MP types effectively loaded EPO-R76E and achieved sustained release, providing detectable levels of EPO-R76E at the injection site in the eye in vivo for at least 28 days. Testing in an in vitro oxidative stress assay and a mouse model of blast-induced indirect traumatic optic neuropathy (bITON) showed that PPS and PLGA MP-mediated delivery of EPO-R76E provided therapeutic protection. While unloaded PLGA MPs inherently increase levels of pro-inflammatory cytokines in the bITON model, drug-free PPS MPs have innate antioxidant properties that provide therapeutic benefit both in vitro and in vivo. Both PLGA and PPS MPs enabled sustained release of EPO-R76E, providing therapeutic benefits including reduction in inflammation and axon degeneration, and preservation of visual function as measured by electroretinogram. The PPS-based MP platform is especially promising for further development, as the delivery system provides inherent antioxidant benefits that can be harnessed to work in complement with EPO-R76E or other drugs for neuroprotection in the setting of traumatic eye injury.
Collapse
Affiliation(s)
- C.R. DeJulius
- Department of Biomedical Engineering, Vanderbilt University, United States
| | - A. Bernardo-Colón
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, United States
| | - S. Naguib
- Department of Ophthalmology & Visual Science, Vanderbilt University School of Medicine, United States
| | - J.R. Backstrom
- Department of Biomedical Engineering, Vanderbilt University, United States,Vanderbilt Eye Institute, Vanderbilt University Medical Center, United States
| | - T. Kavanaugh
- Department of Biomedical Engineering, Vanderbilt University, United States
| | - M.K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, United States
| | - C.L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, United States,Corresponding authors at: Department of Biomedical Engineering, Vanderbilt University, and Vanderbilt Eye Institute, Vanderbilt University Medical Center, United States. (C.L. Duvall), (T.S. Rex)
| | - T.S. Rex
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, United States,Department of Ophthalmology & Visual Science, Vanderbilt University School of Medicine, United States,Corresponding authors at: Department of Biomedical Engineering, Vanderbilt University, and Vanderbilt Eye Institute, Vanderbilt University Medical Center, United States. (C.L. Duvall), (T.S. Rex)
| |
Collapse
|
8
|
Eiger DS, Boldizsar N, Honeycutt CC, Gardner J, Rajagopal S. Biased agonism at chemokine receptors. Cell Signal 2020; 78:109862. [PMID: 33249087 DOI: 10.1016/j.cellsig.2020.109862] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
In the human chemokine system, interactions between the approximately 50 known endogenous chemokine ligands and 20 known chemokine receptors (CKRs) regulate a wide range of cellular functions and biological processes including immune cell activation and homeostasis, development, angiogenesis, and neuromodulation. CKRs are a family of G protein-coupled receptors (GPCR), which represent the most common and versatile class of receptors in the human genome and the targets of approximately one third of all Food and Drug Administration-approved drugs. Chemokines and CKRs bind with significant promiscuity, as most CKRs can be activated by multiple chemokines and most chemokines can activate multiple CKRs. While these ligand-receptor interactions were previously regarded as redundant, it is now appreciated that many chemokine:CKR interactions display biased agonism, the phenomenon in which different ligands binding to the same receptor signal through different pathways with different efficacies, leading to distinct biological effects. Notably, these biased responses can be modulated through changes in ligand, receptor, and or the specific cellular context (system). In this review, we explore the biochemical mechanisms, functional consequences, and therapeutic potential of biased agonism in the chemokine system. An enhanced understanding of biased agonism in the chemokine system may prove transformative in the understanding of the mechanisms and consequences of biased signaling across all GPCR subtypes and aid in the development of biased pharmaceuticals with increased therapeutic efficacy and safer side effect profiles.
Collapse
Affiliation(s)
| | - Noelia Boldizsar
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | | | - Julia Gardner
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
9
|
Smit MJ, Schlecht-Louf G, Neves M, van den Bor J, Penela P, Siderius M, Bachelerie F, Mayor F. The CXCL12/CXCR4/ACKR3 Axis in the Tumor Microenvironment: Signaling, Crosstalk, and Therapeutic Targeting. Annu Rev Pharmacol Toxicol 2020; 61:541-563. [PMID: 32956018 DOI: 10.1146/annurev-pharmtox-010919-023340] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Elevated expression of the chemokine receptors CXCR4 and ACKR3 and of their cognate ligand CXCL12 is detected in a wide range of tumors and the tumor microenvironment (TME). Yet, the molecular mechanisms by which the CXCL12/CXCR4/ACKR3 axis contributes to the pathogenesis are complex and not fully understood. To dissect the role of this axis in cancer, we discuss its ability to impinge on canonical and less conventional signaling networks in different cancer cell types; its bidirectional crosstalk, notably with receptor tyrosine kinase (RTK) and other factors present in the TME; and the infiltration of immune cells that supporttumor progression. We discuss current and emerging avenues that target the CXCL12/CXCR4/ACKR3 axis. Coordinately targeting both RTKs and CXCR4/ACKR3 and/or CXCL12 is an attractive approach to consider in multitargeted cancer therapies. In addition, inhibiting infiltrating immune cells or reactivating the immune system along with modulating the CXCL12/CXCR4/ACKR3 axis in the TME has therapeutic promise.
Collapse
Affiliation(s)
- Martine J Smit
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France
| | - Maria Neves
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France.,Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Jelle van den Bor
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Petronila Penela
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Marco Siderius
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
10
|
Adlere I, Caspar B, Arimont M, Dekkers S, Visser K, Stuijt J, de Graaf C, Stocks M, Kellam B, Briddon S, Wijtmans M, de Esch I, Hill S, Leurs R. Modulators of CXCR4 and CXCR7/ACKR3 Function. Mol Pharmacol 2019; 96:737-752. [DOI: 10.1124/mol.119.117663] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/14/2019] [Indexed: 02/06/2023] Open
|
11
|
Serri C, Frigione M, Ruponen M, Urtti A, Borzacchiello A, Biondi M, Itkonen J, Mayol L. Electron dispersive X-ray spectroscopy and degradation properties of hyaluronic acid decorated microparticles. Colloids Surf B Biointerfaces 2019; 181:896-901. [DOI: 10.1016/j.colsurfb.2019.06.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/28/2019] [Accepted: 06/18/2019] [Indexed: 12/30/2022]
|
12
|
Radhakrishnan K, Vincent A, Joseph RR, Moreno M, Dickescheid A, Agrawal R, Venkatraman S. Hollow Microcapsules as Periocular Drug Depot for Sustained Release of Anti-VEGF Protein. Pharmaceutics 2019; 11:E330. [PMID: 31336771 PMCID: PMC6680760 DOI: 10.3390/pharmaceutics11070330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/03/2019] [Accepted: 07/06/2019] [Indexed: 01/30/2023] Open
Abstract
Diseases affecting the posterior segment of the eye such as age-related macular degeneration and diabetic retinopathy are leading causes of blindness all over the world. The current treatment regimen for such diseases involves repeated intravitreal injections of anti- Vascular Endothelial Growth Factor (VEGF) proteins. This method is highly invasive and can lead to severe complications. In an attempt to develop less invasive alternatives, we propose the use of a controlled release system consisting of anti-VEGF loaded hollow microcapsules that can be administered periocularly to form drug eluting depots on the episcleral surface. The microcapsules with either positive or negative surface charge were prepared by a layer by layer approach and showed pH responsive permeability switching. An ex vivo experiment using porcine sclera indicated positively charged microcapsules remained on the episcleral surface over four days while the negatively charged microcapsules were washed away. These positively charged microcapsules were then loaded with anti-VEGF protein ranibizumab using pH dependent permeability switching and protein release from the microcapsules were studied using an in vitro setup. An ex vivo experiment utilizing porcine sclera demonstrated sustained release of ranibizumab over seven days with zero-order kinetics.
Collapse
Affiliation(s)
- Krishna Radhakrishnan
- NTU-Northwestern Institute for Nanomedicine (NNIN), School of Materials Science and Engineering, Nanyang Technological University (NTU), 50, Nanyang Avenue, Singapore 639798, Singapore.
| | - Anita Vincent
- NTU-Northwestern Institute for Nanomedicine (NNIN), School of Materials Science and Engineering, Nanyang Technological University (NTU), 50, Nanyang Avenue, Singapore 639798, Singapore
| | - Rini Rachel Joseph
- NTU-Northwestern Institute for Nanomedicine (NNIN), School of Materials Science and Engineering, Nanyang Technological University (NTU), 50, Nanyang Avenue, Singapore 639798, Singapore
| | - Miguel Moreno
- NTU-Northwestern Institute for Nanomedicine (NNIN), School of Materials Science and Engineering, Nanyang Technological University (NTU), 50, Nanyang Avenue, Singapore 639798, Singapore
| | - Andreas Dickescheid
- NTU-Northwestern Institute for Nanomedicine (NNIN), School of Materials Science and Engineering, Nanyang Technological University (NTU), 50, Nanyang Avenue, Singapore 639798, Singapore
| | - Rupesh Agrawal
- Tan Tock Seng Hospital, Novena, Singapore 308433, Singapore
| | - Subbu Venkatraman
- NTU-Northwestern Institute for Nanomedicine (NNIN), School of Materials Science and Engineering, Nanyang Technological University (NTU), 50, Nanyang Avenue, Singapore 639798, Singapore.
| |
Collapse
|
13
|
Meng T, Kulkarni V, Simmers R, Brar V, Xu Q. Therapeutic implications of nanomedicine for ocular drug delivery. Drug Discov Today 2019; 24:1524-1538. [PMID: 31102733 DOI: 10.1016/j.drudis.2019.05.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/21/2019] [Accepted: 05/07/2019] [Indexed: 01/01/2023]
Abstract
Delivering therapeutics to the eye is challenging on multiple levels: rapid clearance of eyedrops from the ocular surface requires frequent instillation, which is difficult for patients; transport of drugs across the blood-retinal barrier when drugs are administered systemically, and the cornea when drugs are administered topically, is difficult to achieve; limited drug penetration to the back of the eye owing to the cornea, conjunctiva, sclera and vitreous barriers. Nanomedicine offers many advantages over conventional ophthalmic medications for effective ocular drug delivery because nanomedicine can increase the therapeutic index by overcoming ocular barriers, improving drug-release profiles and reducing potential drug toxicity. In this review, we highlight the therapeutic implications of nanomedicine for ocular drug delivery.
Collapse
Affiliation(s)
- Tuo Meng
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Vineet Kulkarni
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Russell Simmers
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Physics, College of Humanities & Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Vikram Brar
- Department of Ophthalmology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Qingguo Xu
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Ophthalmology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
14
|
Orive G, Santos-Vizcaino E, Pedraz JL, Hernandez RM, Vela Ramirez JE, Dolatshahi-Pirouz A, Khademhosseini A, Peppas NA, Emerich DF. 3D cell-laden polymers to release bioactive products in the eye. Prog Retin Eye Res 2019; 68:67-82. [DOI: 10.1016/j.preteyeres.2018.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/16/2022]
|
15
|
Shatz W, Aaronson J, Yohe S, Kelley RF, Kalia YN. Strategies for modifying drug residence time and ocular bioavailability to decrease treatment frequency for back of the eye diseases. Expert Opin Drug Deliv 2018; 16:43-57. [PMID: 30488721 DOI: 10.1080/17425247.2019.1553953] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Treating posterior eye diseases has become a major area of focus for pharmaceutical and biotechnology companies. Current standard of care for treating posterior eye diseases relies on administration via intravitreal injection. Although effective, this is not without complications and there is great incentive to develop longer-acting therapeutics and/or sustained release delivery systems. Here, we present an overview of emerging technologies for delivery of biologics to the back of the eye. AREAS COVERED Posterior eye diseases, intravitreal injection, age-related macular degeneration, anti-VEGF, ocular pharmacokinetics, novel technologies to extend half-life, in vivo models, translation to the clinic, and hurdles to effective patient care. EXPERT OPINION Posterior eye diseases are a worldwide public health issue. Although anti-VEGF molecules represent a major advance for treating diseases involving choroidal neovascularization, frequent injection can be burdensome for patients and clinicians. There is a need for effective and patient-friendly treatments for posterior eye diseases. Many technologies that enable long-acting delivery to the back of the eye are being evaluated. However, successful development of novel therapies and delivery technologies is hampered by a multitude of factors, including patient education, translatability of in vitro/in vivo preclinical data to the clinic, and regulatory challenges associated with novel technologies.
Collapse
Affiliation(s)
- Whitney Shatz
- a Department of Protein Chemistry , Genentech , South San Francisco , CA , USA.,b School of Pharmaceutical Sciences , University of Geneva & University of Lausanne , Geneva , Switzerland
| | - Jeffrey Aaronson
- c Department of Drug Delivery , Genentech , South San Francisco , CA , USA
| | - Stefan Yohe
- c Department of Drug Delivery , Genentech , South San Francisco , CA , USA
| | - Robert F Kelley
- c Department of Drug Delivery , Genentech , South San Francisco , CA , USA
| | - Yogeshvar N Kalia
- b School of Pharmaceutical Sciences , University of Geneva & University of Lausanne , Geneva , Switzerland
| |
Collapse
|
16
|
Patil MA, Upadhyay AK, Hernandez-Lagunas L, Good R, Carpenter TC, Sucharov CC, Nozik-Grayck E, Kompella UB. Targeted delivery of YSA-functionalized and non-functionalized polymeric nanoparticles to injured pulmonary vasculature. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1059-S1066. [PMID: 30450979 DOI: 10.1080/21691401.2018.1528984] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ephrin type-A receptor 2 (EphA2) is a transmembrane receptor which is upregulated in injured lungs, including those treated with bleomycin. YSA peptide (YSAYPDSVPMMS), a mimic of ephrin ligands, binds to EphA2 receptors on cell surface with high affinity. In this study, we assessed the ability of YSA-functionalized and non-functionalized poly (dl-lactide-co-glycolide) (PLGA) nanoparticles to enhance delivery to bleomycin treated cultured vascular endothelial cells and, in a bleomycin induced lung injury mouse model. Nanoparticles were loaded with a lipophilic fluorescent dye. Human umbilical vein endothelial cells (HUVEC) with or without 2-day bleomycin pretreatment (25 µg/ml) and adult mice with or without intratracheal instillation of bleomycin (0.1 U) were dosed with nanoparticles. Mice received nanoparticles via tail vein injection 4 days after bleomycin treatment. Three days after nanoparticle injection, tissues (lung, heart, kidney, spleen, liver, brain, eyes and whole blood) were harvested and quantified for fluorescence using IVIS imaging. Mean particle uptake increased with time and concentration for both types of particles in HUVEC, with the uptake being higher for YSA-functionalized nanoparticles. Bleomycin treatment increased the 3-h uptake of both types of nanoparticles in HUVEC by about two-fold, with the YSA-functionalized nanoparticle uptake being 1.66-fold compared to non-functionalized nanoparticles (p < .05). In mice, bleomycin injury resulted in 2.3- and 4.7-fold increase in the lung levels of non-functionalized and YSA-functionalized nanoparticles (p < .05), respectively, although the differences between the two particle types were not significant. In conclusion, PLGA nanoparticle delivery to cultured vascular endothelial cells and mouse lungs in vivo is higher following bleomycin treatment, with the delivery tending to be higher for YSA functionalized nanoparticles.
Collapse
Affiliation(s)
- Madhoosudan A Patil
- a Department of Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Arun K Upadhyay
- a Department of Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Laura Hernandez-Lagunas
- b Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine , University of Colorado Denver , Denver , CO , USA
| | - Ryan Good
- b Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine , University of Colorado Denver , Denver , CO , USA
| | - Todd C Carpenter
- c Division of Pediatric Critical Care Medicine, Department of Pediatrics , University of Colorado School of Medicine , Aurora , CO , USA
| | - Carmen C Sucharov
- d Division of Cardiology, Department of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Eva Nozik-Grayck
- b Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine , University of Colorado Denver , Denver , CO , USA.,c Division of Pediatric Critical Care Medicine, Department of Pediatrics , University of Colorado School of Medicine , Aurora , CO , USA
| | - Uday B Kompella
- a Department of Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,e Department of Ophthalmology , University of Colorado Anschutz Medical Campus , Aurora , Colorado.,f Department of Bioengineering , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,g Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| |
Collapse
|
17
|
Mezu-Ndubuisi OJ, Wang Y, Schoephoerster J, Falero-Perez J, Zaitoun IS, Sheibani N, Gong S. Intravitreal Delivery of VEGF-A 165-loaded PLGA Microparticles Reduces Retinal Vaso-Obliteration in an In Vivo Mouse Model of Retinopathy of Prematurity. Curr Eye Res 2018; 44:275-286. [PMID: 30383455 DOI: 10.1080/02713683.2018.1542736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE Retinopathy of prematurity (ROP) is a condition of abnormal retinal vascularization with reduced levels of vascular endothelial growth factor (VEGF) causing vaso-obliteration (Phase I), followed by abnormal neovascularization from increased VEGF (Phase II). We hypothesized that intravitreal pro-angiogenic VEGF-A in microparticle form would promote earlier retinal revascularization in an oxygen-induced ischemic retinopathy (OIR) mouse model. MATERIALS AND METHODS Wildtype mice (39) were exposed to 77% oxygen from postnatal day 7 (P7) to P12. VEGF-A165-loaded poly(lactic-co-glycolic acid) (PLGA) (n = 15) or empty PLGA (n = 14) microparticles were fabricated using a water-in-oil-in-water double emulsion method, and injected intravitreally at P13 into mice right eyes (RE). Left eyes (LE) were untreated. At P20, after retinal fluorescein angiography, vascular parameters were quantified. Retinal VEGF levels at P13 and flatmounts at P20 were performed separately. RESULTS VEGF-A165-loaded microparticles had a mean diameter of 4.2 μm. with a loading level of 8.6 weight.%. Retinal avascular area was reduced in VEGF-treated RE (39.5 ± 9.0%) compared to untreated LE (52.6 ± 6.1%, p < 0.0001) or empty microparticle-treated RE (p < 0.001) and untreated LEs (p = 0.001). Retinal arteries in VEGF-treated RE were less tortuous than untreated LE (1.08 ± 0.05 vs. 1.18 ± 0.08, p < 0.001) or empty-microparticles-treated RE (p = 0.02). Retinal arterial tortuosity was similar in the LE of VEGF and empty microparticle-treated mice (P > 0.05). Retinal vein width was similar in VEGF-treated and empty microparticle-treated RE (P > 0.9), which were each less dilated than their contralateral LE (p < 0.01). VEGF levels were higher in P13 OIR mice than RA mice (p < 0.0001). Retinal flatmounts showed vaso-obliteration and neovascularization. CONCLUSIONS Endogenous retinal VEGF is suppressed in OIR mice. Exogenous intravitreal VEGF-A165-loaded microparticles in OIR mice reduced retinal vaso-obliteration and accelerated recovery from vein dilation and arterial tortuosity. This may be beneficial in preventing Phase II ROP without systemic effects.
Collapse
Affiliation(s)
- Olachi J Mezu-Ndubuisi
- a Department of Pediatrics , University of Wisconsin-Madison , Madison , WI , USA.,b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA
| | - Yuyuan Wang
- c Department of Materials Science and Engineering and Wisconsin Institute for Discovery , University of Wisconsin-Madison , Madison , WI , USA
| | - Jamee Schoephoerster
- a Department of Pediatrics , University of Wisconsin-Madison , Madison , WI , USA
| | - Juliana Falero-Perez
- b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA
| | - Ismail S Zaitoun
- b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA
| | - Nader Sheibani
- b Department of Ophthalmology and Visual Sciences , University of Wisconsin-Madison , Madison , WI , USA.,d Department of Biomedical Engineering , University of Wisconsin-Madison , Madison , WI , USA
| | - Shaoqin Gong
- c Department of Materials Science and Engineering and Wisconsin Institute for Discovery , University of Wisconsin-Madison , Madison , WI , USA.,d Department of Biomedical Engineering , University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
18
|
Anti‐Flt1 peptide and cyanine‐conjugated gold nanoparticles for the concurrent antiangiogenic and endothelial cell proton treatment. J Biomed Mater Res B Appl Biomater 2018; 107:1272-1283. [DOI: 10.1002/jbm.b.34220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/09/2018] [Accepted: 08/02/2018] [Indexed: 01/03/2023]
|
19
|
Rafati A, Ebadi A, Bavafa S, Nowroozi A. Kinetic study, structural analysis and computational investigation of novel xerogel based on drug-PEG/SiO2 for controlled release of enrofloxacin. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2018.06.104] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
20
|
Polymer-based carriers for ophthalmic drug delivery. J Control Release 2018; 285:106-141. [DOI: 10.1016/j.jconrel.2018.06.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/23/2018] [Accepted: 06/25/2018] [Indexed: 12/22/2022]
|
21
|
Abd AJ, Kanwar RK, Pathak YV, Al Mohammedawi M, Kanwar JR. Nanomedicine-Based Delivery to the Posterior Segment of the Eye: Brighter Tomorrow. DRUG DELIVERY FOR THE RETINA AND POSTERIOR SEGMENT DISEASE 2018:195-212. [DOI: 10.1007/978-3-319-95807-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
22
|
Joseph RR, Tan DWN, Ramon MRM, Natarajan JV, Agrawal R, Wong TT, Venkatraman SS. Characterization of liposomal carriers for the trans-scleral transport of Ranibizumab. Sci Rep 2017; 7:16803. [PMID: 29196745 PMCID: PMC5711922 DOI: 10.1038/s41598-017-16791-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/13/2017] [Indexed: 01/04/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness in the modern world. The standard treatment regimen for neovascular AMD is the monthly/bimonthly intravitreal injection of anti-VEGF agents such as ranibizumab or aflibercept. However, these repeated invasive injections can lead to sight-threatening complications. Sustained delivery by encapsulation of the drug in carriers is a way to reduce the frequency of these injections. Liposomes are biocompatible, non-toxic vesicular nanocarriers, which can be used to encapsulate therapeutic agents to provide sustained release. The protein encapsulation was performed by a modified dehydration-rehydration (DRV) method. The liposomes formed were characterized for size, zeta potential, encapsulation efficiency, stability, in vitro release, and ex vivo release profiles. In addition, the localization of the liposomes themselves was studied ex vivo. Entrapment-efficiency of ranibizumab into 100-nm liposomes varied from 14.7 to 57.0%. Negatively-charged liposomes prepared from DPPC-DPPG were found to have the slowest release with a low initial burst release compared to the rest of liposomal formulations. The ex vivo protein release was found to slower than the in vitro protein release for all samples. In conclusion, the DPPC-DPPG liposomes significantly improved the encapsulation and release profile of ranibizumab.
Collapse
Affiliation(s)
- Rini Rachel Joseph
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Dulcia Wei Ni Tan
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Moreno Raja Miguel Ramon
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jayaganesh V Natarajan
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Rupesh Agrawal
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tina T Wong
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore.
- Singapore National Eye Centre, Singapore, Singapore.
- Ocular Drug Delivery Group, Singapore Eye Research Institute, Singapore, Singapore.
| | - Subbu S Venkatraman
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
23
|
Assessment of different polymers and drug loads for fused deposition modeling of drug loaded implants. Eur J Pharm Biopharm 2017; 115:84-93. [PMID: 28232106 DOI: 10.1016/j.ejpb.2017.02.014] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/19/2017] [Accepted: 02/17/2017] [Indexed: 11/20/2022]
Abstract
The 3D printing technique of fused deposition modeling® (FDM) has lately come into focus as a potential fabrication technique for pharmaceutical dosage forms and medical devices that allows the preparation of delivery systems with nearly any shape. This is particular promising for implants administered at application sites with a high anatomical variability where an individual shape adaption appears reasonable. In this work different polymers (Eudragit®RS, polycaprolactone (PCL), poly(l-lactide) (PLLA) and ethyl cellulose (EC)) were evaluated with respect to their suitability for FDM of drug loaded implants and their drug release behaviour was evaluated. The fluorescent dye quinine was used as a model drug to visualize drug distribution in filaments and implants. Quinine loaded filaments were produced by solvent casting and subsequent hot melt extrusion (HME) and model implants were printed as hollow cylinders using a standard FDM printer. Parameters were found at which model implants (hollow cylinders, outer diameter 4-5mm, height 3mm) could be produced from all tested polymers. The drug release which was examined by incubation of the printed implants in phosphate buffered saline solution (PBS) pH 7.4 was highly dependent on the used polymer. The fastest relative drug release of approximately 76% in 51days was observed for PCL and the lowest for Eudragit®RS and EC with less than 5% of quinine release in 78 and 100days, respectively. For PCL further filaments were prepared with different quinine loads ranging from 2.5% to 25% and thermal analysis proved the presence of a solid dispersion of quinine in the polymer for all tested concentrations. Increasing the drug load also increased the overall percentage of drug released to the medium since nearly the same absolute amount of quinine remained trapped in PCL at the end of drug release studies. This knowledge is valuable for future developments of printed implants with a desired drug release profile that might be controlled by the choice of the polymer and the drug load.
Collapse
|
24
|
Joseph RR, Venkatraman SS. Drug delivery to the eye: what benefits do nanocarriers offer? Nanomedicine (Lond) 2017; 12:683-702. [PMID: 28186436 DOI: 10.2217/nnm-2016-0379] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ocular drug delivery has seen several advances in the past few decades, with respect to new drugs, improved formulations, targeted delivery, as well as exploration of new routes of drug administration. New materials have been explored for encasing existing drugs, which can enhance treatment by increasing bioavailability, decreasing toxicity, providing better tissue adherence, targeted delivery as well as increased duration of action. The challenges and requirements are different for the anterior and posterior ocular segments. This review summarizes the recent advances in sustained ocular therapy, both to the anterior and posterior segments, which have been made possible, thanks to nanotechnology. We also discuss the distribution and fate of these nanocarriers themselves, postadministration, as well as clearance from ocular tissues.
Collapse
Affiliation(s)
- Rini Rachel Joseph
- School of Materials Science & Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Subbu S Venkatraman
- School of Materials Science & Engineering, Nanyang Technological University, Singapore 639798, Singapore.,NTU-Northwestern Institute for Nanomedicine, School of Materials Science & Engineering (MSE), Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
25
|
Del Amo EM, Rimpelä AK, Heikkinen E, Kari OK, Ramsay E, Lajunen T, Schmitt M, Pelkonen L, Bhattacharya M, Richardson D, Subrizi A, Turunen T, Reinisalo M, Itkonen J, Toropainen E, Casteleijn M, Kidron H, Antopolsky M, Vellonen KS, Ruponen M, Urtti A. Pharmacokinetic aspects of retinal drug delivery. Prog Retin Eye Res 2016; 57:134-185. [PMID: 28028001 DOI: 10.1016/j.preteyeres.2016.12.001] [Citation(s) in RCA: 416] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/25/2016] [Accepted: 12/01/2016] [Indexed: 12/14/2022]
Abstract
Drug delivery to the posterior eye segment is an important challenge in ophthalmology, because many diseases affect the retina and choroid leading to impaired vision or blindness. Currently, intravitreal injections are the method of choice to administer drugs to the retina, but this approach is applicable only in selected cases (e.g. anti-VEGF antibodies and soluble receptors). There are two basic approaches that can be adopted to improve retinal drug delivery: prolonged and/or retina targeted delivery of intravitreal drugs and use of other routes of drug administration, such as periocular, suprachoroidal, sub-retinal, systemic, or topical. Properties of the administration route, drug and delivery system determine the efficacy and safety of these approaches. Pharmacokinetic and pharmacodynamic factors determine the required dosing rates and doses that are needed for drug action. In addition, tolerability factors limit the use of many materials in ocular drug delivery. This review article provides a critical discussion of retinal drug delivery, particularly from the pharmacokinetic point of view. This article does not include an extensive review of drug delivery technologies, because they have already been reviewed several times recently. Instead, we aim to provide a systematic and quantitative view on the pharmacokinetic factors in drug delivery to the posterior eye segment. This review is based on the literature and unpublished data from the authors' laboratory.
Collapse
Affiliation(s)
- Eva M Del Amo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Anna-Kaisa Rimpelä
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Emma Heikkinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Otto K Kari
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Eva Ramsay
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tatu Lajunen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Mechthild Schmitt
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Laura Pelkonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Madhushree Bhattacharya
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Dominique Richardson
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tiina Turunen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Jaakko Itkonen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Elisa Toropainen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marco Casteleijn
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Maxim Antopolsky
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | | | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Arto Urtti
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
26
|
Stratton S, Shelke NB, Hoshino K, Rudraiah S, Kumbar SG. Bioactive polymeric scaffolds for tissue engineering. Bioact Mater 2016; 1:93-108. [PMID: 28653043 PMCID: PMC5482547 DOI: 10.1016/j.bioactmat.2016.11.001] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/27/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022] Open
Abstract
A variety of engineered scaffolds have been created for tissue engineering using polymers, ceramics and their composites. Biomimicry has been adopted for majority of the three-dimensional (3D) scaffold design both in terms of physicochemical properties, as well as bioactivity for superior tissue regeneration. Scaffolds fabricated via salt leaching, particle sintering, hydrogels and lithography have been successful in promoting cell growth in vitro and tissue regeneration in vivo. Scaffold systems derived from decellularization of whole organs or tissues has been popular due to their assured biocompatibility and bioactivity. Traditional scaffold fabrication techniques often failed to create intricate structures with greater resolution, not reproducible and involved multiple steps. The 3D printing technology overcome several limitations of the traditional techniques and made it easier to adopt several thermoplastics and hydrogels to create micro-nanostructured scaffolds and devices for tissue engineering and drug delivery. This review highlights scaffold fabrication methodologies with a focus on optimizing scaffold performance through the matrix pores, bioactivity and degradation rate to enable tissue regeneration. Review highlights few examples of bioactive scaffold mediated nerve, muscle, tendon/ligament and bone regeneration. Regardless of the efforts required for optimization, a shift in 3D scaffold uses from the laboratory into everyday life is expected in the near future as some of the methods discussed in this review become more streamlined.
Collapse
Affiliation(s)
- Scott Stratton
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Namdev B. Shelke
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, USA
- Institute for Regenerative Engineering, UConn Health, Farmington, CT, USA
| | - Kazunori Hoshino
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Swetha Rudraiah
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Saint Joseph, Hartford, CT, 06103, USA
| | - Sangamesh G. Kumbar
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, USA
- Institute for Regenerative Engineering, UConn Health, Farmington, CT, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
27
|
Käsdorf BT, Arends F, Lieleg O. Diffusion Regulation in the Vitreous Humor. Biophys J 2016; 109:2171-81. [PMID: 26588575 DOI: 10.1016/j.bpj.2015.10.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/28/2015] [Accepted: 10/01/2015] [Indexed: 02/02/2023] Open
Abstract
The efficient treatment of many ocular diseases depends on the rapid diffusive distribution of solutes such as drugs or drug delivery vehicles through the vitreous humor. However, this multicomponent hydrogel possesses selective permeability properties, which allow for the diffusion of certain molecules and particles, whereas others are immobilized. In this study, we perform an interspecies comparison showing that the selective permeability properties of the vitreous are conserved across several mammalian species. We identify the polyanionic glycosaminoglycans hyaluronic acid and heparan sulfate as two key macromolecules that establish this selective permeability. We show that electrostatic interactions between the polyanionic macromolecules and diffusing solutes can be weakened by charge screening or enzymatic glycosaminoglycan digestion. Furthermore, molecule penetration into the vitreous is also charge-dependent and only efficient as long as the net charge of the molecule does not exceed a certain threshold.
Collapse
Affiliation(s)
- Benjamin Tillmann Käsdorf
- Institute of Medical Engineering (IMETUM), Technische Universität München, Garching, Germany; Department of Mechanical Engineering, Technische Universität München, Garching, Germany
| | - Fabienna Arends
- Institute of Medical Engineering (IMETUM), Technische Universität München, Garching, Germany; Department of Mechanical Engineering, Technische Universität München, Garching, Germany
| | - Oliver Lieleg
- Institute of Medical Engineering (IMETUM), Technische Universität München, Garching, Germany; Department of Mechanical Engineering, Technische Universität München, Garching, Germany.
| |
Collapse
|
28
|
Villegas VM, Aranguren LA, Kovach JL, Schwartz SG, Flynn HW. Current advances in the treatment of neovascular age-related macular degeneration. Expert Opin Drug Deliv 2016; 14:273-282. [PMID: 27434329 DOI: 10.1080/17425247.2016.1213240] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is the most common cause of permanent central visual acuity loss in persons over 65 years of age in industrialized nations. Today, intravitreal vascular endothelial growth factor (VEGF) inhibitors are the mainstay of treatment worldwide. Areas covered: The following review covers the current treatments and challenges of wet AMD management. It also covers emerging therapies including radiation, latest generation anti-VEGF agents, and combination therapies. Expert opinion: Current neovascular AMD therapy is aimed at decreasing the VEGF effect at the choroidal neovascularization (CNV) complex. The most important existing challenges in the treatment of neovascular AMD are improving visual outcomes, decreasing the treatment burden, and minimizing geographic atrophy. Clinicians are using many treatment strategies to minimize intravitreal injections without sacrificing visual outcomes. Combination of anti-VEGF therapy with other previously available treatments that target a different pathophysiological mechanism may be a reasonable clinical strategy to minimize intravitreal injections. Many exciting novel drugs that target newly discovered pathways associated with CNV development and progression hold clinical promise. The results of ongoing randomized clinical trials will answer the important concerns surrounding new drugs and delivery devices: safety and visual outcomes.
Collapse
Affiliation(s)
- Victor M Villegas
- a Department of Ophthalmology, Bascom Palmer Eye Institute , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Luis A Aranguren
- a Department of Ophthalmology, Bascom Palmer Eye Institute , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Jaclyn L Kovach
- a Department of Ophthalmology, Bascom Palmer Eye Institute , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Stephen G Schwartz
- a Department of Ophthalmology, Bascom Palmer Eye Institute , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Harry W Flynn
- a Department of Ophthalmology, Bascom Palmer Eye Institute , University of Miami Miller School of Medicine , Miami , FL , USA
| |
Collapse
|
29
|
Elsaid N, Jackson TL, Elsaid Z, Alqathama A, Somavarapu S. PLGA Microparticles Entrapping Chitosan-Based Nanoparticles for the Ocular Delivery of Ranibizumab. Mol Pharm 2016; 13:2923-40. [PMID: 27286558 DOI: 10.1021/acs.molpharmaceut.6b00335] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of certified vision loss worldwide. The standard treatment for neovascular AMD involves repeated intravitreal injections of therapeutic proteins directed against vascular endothelial growth factor, such as ranibizumab. Biodegradable polymers, such as poly(lactic-co-glycolic acid) (PLGA), form delivery vehicles which can be used to treat posterior segment eye diseases, but suffer from poor protein loading and release. This work describes a "system-within-system", PLGA microparticles incorporating chitosan-based nanoparticles, for improved loading and sustained intravitreal delivery of ranibizumab. Chitosan-N-acetyl-l-cysteine (CNAC) was synthesized and its synthesis confirmed using FT-IR and (1)H NMR. Chitosan-based nanoparticles composed of CNAC, CNAC/tripolyphosphate (CNAC/TPP), chitosan, chitosan/TPP (chit/TPP), or chit/TPP-hyaluronic acid (chit/TPP-HA) were incorporated in PLGA microparticles using a modified w/o/w double emulsion method. Nanoparticles and final nanoparticles-within-microparticles were characterized for their protein-nanoparticle interaction, size, zeta potential, morphology, protein loading, stability, in vitro release, in vivo antiangiogenic activity, and effects on cell viability. The prepared nanoparticles were 17-350 nm in size and had zeta potentials of -1.4 to +12 mV. Microscopic imaging revealed spherical nanoparticles on the surface of PLGA microparticles for preparations containing chit/TPP, CNAC, and CNAC/TPP. Ranibizumab entrapment efficiency in the preparations varied between 13 and 69% and was highest for the PLGA microparticles containing CNAC nanoparticles. This preparation also showed the slowest release with no initial burst release compared to all other preparations. Incorporation of TPP to this formulation increased the rate of protein release and reduced entrapment efficiency. PLGA microparticles containing chit/TPP-HA showed the fastest and near-complete release of ranibizumab. All of the prepared empty particles showed no effect on cell viability up to a concentration of 12.5 mg/mL. Ranibizumab released from all preparations maintained its structural integrity and in vitro activity. The chit/TPP-HA preparation enhanced antiangiogenic activity and may provide a potential biocompatible platform for enhanced antiangiogenic activity in combination with ranibizumab. In conclusion, the PLGA microparticles containing CNAC nanoparticles showed significantly improved ranibizumab loading and release profile. This novel drug delivery system may have potential for improved intravitreal delivery of therapeutic proteins, thereby reducing the frequency, risk, and cost of burdensome intravitreal injections.
Collapse
Affiliation(s)
- Naba Elsaid
- University of Hertfordshire , Hatfield, United Kingdom
| | | | - Zeeneh Elsaid
- University College London School of Pharmacy , London, United Kingdom
| | | | | |
Collapse
|
30
|
Bora M, Mundargi RC, Chee Y, Wong TT, Venkatraman SS. 5-Flurouracil microencapsulation and impregnation in hyaluronic acid hydrogel as composite drug delivery system for ocular fibrosis. COGENT MEDICINE 2016. [DOI: 10.1080/2331205x.2016.1182108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Meghali Bora
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Raghavendra C. Mundargi
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - YongDe Chee
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Tina T.L. Wong
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, 11 Third Hospital Avenue, Singapore 168751, Singapore
| | - Subbu S. Venkatraman
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
31
|
Shen HH, Chan EC, Lee JH, Bee YS, Lin TW, Dusting GJ, Liu GS. Nanocarriers for treatment of ocular neovascularization in the back of the eye: new vehicles for ophthalmic drug delivery. Nanomedicine (Lond) 2015; 10:2093-107. [PMID: 26096379 DOI: 10.2217/nnm.15.47] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pathologic neovascularization of the retina is a major cause of substantial and irreversible loss of vision. Drugs are difficult to deliver to the lesions in the back of the eye and this is a major obstacle for the therapeutics. Current pharmacological approach involves an intravitreal injection of anti-VEGF agents to prevent aberrant growth of blood vessels, but it has limitations including therapeutic efficacy and side-effects associated with systemic exposure and invasive surgery. Nanotechnology provides novel opportunities to overcome the limitations of conventional delivery system to reach the back of the eye through fabrication of nanostructures capable of encapsulating and delivering small molecules. This review article introduces various forms of nanocarrier that can be adopted by ocular drug delivery systems to improve current therapy. The application of nanotechnology in medicine brings new hope for ocular drug delivery in the back of the eye to manage the major causes of blindness associated with ocular neovascularization.
Collapse
Affiliation(s)
- Hsin-Hui Shen
- Department of Microbiology, Monash University, Clayton, Melbourne, VIC, Australia
| | - Elsa C Chan
- Centre for Eye Research Australia, East Melbourne, VIC, Australia.,Department of Ophthalmology, University of Melbourne, East Melbourne, VIC, Australia
| | - Jia Hui Lee
- Centre for Eye Research Australia, East Melbourne, VIC, Australia
| | - Youn-Shen Bee
- Department of Ophthalmology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Yuh-Ing Junior College of Health Care & Management, Kaohsiung, Taiwan.,National Defense Medical Center, Taipei, Taiwan
| | - Tsung-Wu Lin
- Department of Chemistry, Tunghai University, Taichung City, Taiwan
| | - Gregory J Dusting
- Centre for Eye Research Australia, East Melbourne, VIC, Australia.,Department of Ophthalmology, University of Melbourne, East Melbourne, VIC, Australia
| | - Guei-Sheung Liu
- Centre for Eye Research Australia, East Melbourne, VIC, Australia.,Department of Ophthalmology, University of Melbourne, East Melbourne, VIC, Australia
| |
Collapse
|
32
|
Zabihi F, Yang M, Leng Y, Zhao Y. PLGA–HPMC nanoparticles prepared by a modified supercritical anti-solvent technique for the controlled release of insulin. J Supercrit Fluids 2015. [DOI: 10.1016/j.supflu.2015.01.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
33
|
Pan Q, Xu Q, Boylan NJ, Lamb NW, Emmert DG, Yang JC, Tang L, Heflin T, Alwadani S, Eberhart CG, Stark WJ, Hanes J. Corticosteroid-loaded biodegradable nanoparticles for prevention of corneal allograft rejection in rats. J Control Release 2015; 201:32-40. [PMID: 25576786 DOI: 10.1016/j.jconrel.2015.01.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 12/20/2014] [Accepted: 01/05/2015] [Indexed: 10/24/2022]
Abstract
Immunologic graft rejection is one of the main causes of short and long-term graft failure in corneal transplantation. Steroids are the most commonly used immunosuppressive agents for postoperative management and prevention of corneal graft rejection. However, steroids delivered in eye drops are rapidly cleared from the surface of the eye, so the required frequency of dosing for corneal graft rejection management can be as high as once every 2h. Additionally, these eye drops are often prescribed for daily use for 1 year or longer, which can result in poor patient compliance and steroid-related side effects. Here, we report a biodegradable nanoparticle system composed of Generally Regarded as Safe (GRAS) materials that can provide sustained release of corticosteroids to prevent corneal graft rejection following subconjunctival injection provided initially during transplant surgery. Poly(lactic-co-glycolic acid) (PLGA) nanoparticles containing dexamethasone sodium phosphate (DSP) exhibited a size of 200 nm, 8 wt.% drug loading, and sustained drug release over 15 days in vitro under sink conditions. DSP-loaded nanoparticles provided sustained ocular drug levels for at least 7 days after subconjunctival administration in rats, and prevented corneal allograft rejection over the entire 9-week study when administered weekly. In contrast, control treatment groups that received weekly injections of either placebo nanoparticles, saline, or DSP in solution demonstrated corneal graft rejection accompanied by severe corneal edema, neovascularization and opacity that occurred in ≤ 4 weeks. Local controlled release of corticosteroids may reduce the rate of corneal graft rejection, perhaps especially in the days immediately following surgery when risk of rejection is highest and when typical steroid eye drop administration requirements are particularly onerous.
Collapse
Affiliation(s)
- Qing Pan
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, Zhejiang Provicial People's Hospital, Hangzhou, China
| | - Qingguo Xu
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - Nicholas J Boylan
- Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Nicholas W Lamb
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - David G Emmert
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - Jeh-Chang Yang
- Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Li Tang
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, The affiliated hospital of Guiyang medical college, The 28th Guiyi Street, Guiyang, Guizhou, 550004, PR China
| | - Tom Heflin
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - Saeed Alwadani
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Ophthalmology, King Saud University School of Medicine, Riyadh, Saudi Arabia
| | - Charles G Eberhart
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | - Walter J Stark
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA.
| | - Justin Hanes
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
34
|
Shelke NB, James R, Laurencin CT, Kumbar SG. Polysaccharide biomaterials for drug delivery and regenerative engineering. POLYM ADVAN TECHNOL 2014. [DOI: 10.1002/pat.3266] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Namdev B. Shelke
- Institute for Regenerative Engineering; University of Connecticut Health Center; Farmington CT 06030 USA
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences; University of Connecticut Health Center; Farmington CT 06030 USA
- Department of Orthopaedic Surgery; University of Connecticut Health Center; Farmington CT 06030 USA
| | - Roshan James
- Institute for Regenerative Engineering; University of Connecticut Health Center; Farmington CT 06030 USA
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences; University of Connecticut Health Center; Farmington CT 06030 USA
- Department of Orthopaedic Surgery; University of Connecticut Health Center; Farmington CT 06030 USA
| | - Cato T. Laurencin
- Institute for Regenerative Engineering; University of Connecticut Health Center; Farmington CT 06030 USA
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences; University of Connecticut Health Center; Farmington CT 06030 USA
- Department of Orthopaedic Surgery; University of Connecticut Health Center; Farmington CT 06030 USA
- Departments of Materials and Biomedical Engineering; University of Connecticut; Storrs CT 06269 USA
| | - Sangamesh G. Kumbar
- Institute for Regenerative Engineering; University of Connecticut Health Center; Farmington CT 06030 USA
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical, and Engineering Sciences; University of Connecticut Health Center; Farmington CT 06030 USA
- Department of Orthopaedic Surgery; University of Connecticut Health Center; Farmington CT 06030 USA
- Departments of Materials and Biomedical Engineering; University of Connecticut; Storrs CT 06269 USA
| |
Collapse
|
35
|
Xu Q, Kambhampati SP, Kannan RM. Nanotechnology approaches for ocular drug delivery. Middle East Afr J Ophthalmol 2014; 20:26-37. [PMID: 23580849 PMCID: PMC3617524 DOI: 10.4103/0974-9233.106384] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Blindness is a major health concern worldwide that has a powerful impact on afflicted individuals and their families, and is associated with enormous socio-economical consequences. The Middle East is heavily impacted by blindness, and the problem there is augmented by an increasing incidence of diabetes in the population. An appropriate drug/gene delivery system that can sustain and deliver therapeutics to the target tissues and cells is a key need for ocular therapies. The application of nanotechnology in medicine is undergoing rapid progress, and the recent developments in nanomedicine-based therapeutic approaches may bring significant benefits to address the leading causes of blindness associated with cataract, glaucoma, diabetic retinopathy and retinal degeneration. In this brief review, we highlight some promising nanomedicine-based therapeutic approaches for drug and gene delivery to the anterior and posterior segments.
Collapse
Affiliation(s)
- Qingguo Xu
- Department of Ophthalmology, Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
36
|
Panda JJ, Yandrapu S, Kadam RS, Chauhan VS, Kompella UB. Self-assembled phenylalanine-α,β-dehydrophenylalanine nanotubes for sustained intravitreal delivery of a multi-targeted tyrosine kinase inhibitor. J Control Release 2013; 172:1151-60. [PMID: 24075925 PMCID: PMC6349364 DOI: 10.1016/j.jconrel.2013.09.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 12/22/2022]
Abstract
Current standard of care for sustained back of the eye drug delivery is surgical placement or injection of large, slow release implants using a relatively large 22 gauge needle. We designed novel dipeptide (phenylalanine-α,β-dehydrophenylalanine; Phe-∆Phe) based nanotubes with a diameter of ~15-30 nm and a length of ~1500 nm that could be injected with a 33 gauge needle for sustained intravitreal delivery of pazopanib, a multi-targeted tyrosine kinase inhibitor. The drug could be loaded during nanotube assembly or post-loaded after nanotube formation, with the former being more efficient at 25% w/w pazopanib loading and ~55% loading efficiency. Plain and peptide loaded nanotube were non-cytotoxic to retinal pigment epithelial cells even at a concentration of 200 μg/ml. Following intravitreal injection of fluorescently labeled nanotubes using a 33 gauge needle in a rat model, the nanotube persistence and drug delivery were monitored using noninvasive fluorophotometry, electron microscopy and mass spectrometry analysis. Nanotubes persisted in the vitreous humor during the 15 days study and pazopanib levels in the vitreous humor, retina, and choroid-RPE at the end of the study were 4.5, 5, and 2.5-folds higher, respectively, compared to the plain drug. Thus, Phe-∆Phe nanotubes allow intravitreal injections with a small gauge needle and sustain drug delivery.
Collapse
Affiliation(s)
- Jiban J. Panda
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India 110067
- Nanomedicine and Drug Delivery Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sarath Yandrapu
- Nanomedicine and Drug Delivery Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Rajendra S. Kadam
- Nanomedicine and Drug Delivery Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Virander S. Chauhan
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India 110067
| | - Uday B. Kompella
- Nanomedicine and Drug Delivery Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Ophthlamology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
37
|
Schwartz SG, Scott IU, Flynn HW, Stewart MW. Drug delivery techniques for treating age-related macular degeneration. Expert Opin Drug Deliv 2013; 11:61-8. [DOI: 10.1517/17425247.2013.859135] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
38
|
Wang J, Jiang A, Joshi M, Christoforidis J. Drug delivery implants in the treatment of vitreous inflammation. Mediators Inflamm 2013; 2013:780634. [PMID: 24191132 PMCID: PMC3804444 DOI: 10.1155/2013/780634] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/06/2013] [Indexed: 11/25/2022] Open
Abstract
The eye is a model organ for the local delivery of therapeutics. This proves beneficial when treating vitreous inflammation and other ophthalmic pathologies. The chronicity of certain diseases, however, limits the effectiveness of locally administered drugs. To maintain such treatments often requires frequent office visits and can result in increased risk of infection and toxicity to the patient. This paper focuses on the implantable devices and particulate drug delivery systems that are currently being implemented and investigated to overcome these challenges. Implants currently on the market or undergoing clinical trials include those made of nonbiodegradable polymers, containing ganciclovir, fluocinolone acetonide, triamcinolone acetonide, and ranibizumab, and biodegradable polymers, containing dexamethasone, triamcinolone acetonide, and ranibizumab. Investigational intravitreal implants and particulate drug delivery systems, such as nanoparticles, microparticles, and liposomes, are also explored in this review article.
Collapse
Affiliation(s)
- Jillian Wang
- Department of Ophthalmology, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Suite 5000, Columbus, OH 43212, USA
| | - Angela Jiang
- Department of Ophthalmology, The Ohio State University Wexner Medical Center, 915 Olentangy River Road, Suite 5000, Columbus, OH 43212, USA
| | - Malav Joshi
- Department of Ophthalmology & Visual Science, The University of Arizona Medical Center, 655 N. Alvernon Way, Suite 108, Tucson, AZ 85711, USA
| | - John Christoforidis
- Department of Ophthalmology & Visual Science, The University of Arizona Medical Center, 655 N. Alvernon Way, Suite 108, Tucson, AZ 85711, USA
| |
Collapse
|
39
|
Shmueli RB, Ohnaka M, Miki A, Pandey NB, Lima e Silva R, Koskimaki JE, Kim J, Popel AS, Campochiaro PA, Green JJ. Long-term suppression of ocular neovascularization by intraocular injection of biodegradable polymeric particles containing a serpin-derived peptide. Biomaterials 2013; 34:7544-51. [PMID: 23849876 DOI: 10.1016/j.biomaterials.2013.06.044] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/23/2013] [Indexed: 12/27/2022]
Abstract
Aberrant angiogenesis can cause or contribute to a number of diseases such as neovascular age-related macular degeneration (NVAMD). While current NVAMD treatments target angiogenesis, these treatments are not effective for all patients and also require frequent intravitreal injections. New agents and delivery systems to treat NVAMD could be beneficial to many patients. We have recently developed a serpin-derived peptide as an anti-angiogenic agent. Here, this peptide is investigated for activity in human retinal endothelial cells in vitro and for reducing angiogenesis in a laser-induced choroidal neovascularization mouse model of NVAMD in vivo. While frequent intravitreal injections can be tolerated clinically, reducing the number of injections can improve patient compliance, safety, and outcomes. To achieve this goal, and to maximize the in vivo activity of injected peptide, we have developed biodegradable polymers and controlled release particle formulations to extend anti-angiogenic therapy. To create these devices, the anionic peptides are first self-assembled into nanoparticles using a biodegradable cationic polymer and then as a second step, these nanoparticles are encapsulated into biodegradable poly(lactic-co-glycolic acid) (PLGA) microparticles. In situ, these particles show approximately zero-order, linear release of the anionic peptide over 200 days. These particles are made of safe, hydrolytically degradable polymers and have low endotoxin. Long-term in vivo experiments in the laser-induced neovascularization model for NVAMD show that these peptide-releasing particles decrease angiogenesis for at least fourteen weeks in vivo following a single particle dose and therefore are a promising treatment strategy for NVAMD.
Collapse
Affiliation(s)
- Ron B Shmueli
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Xu Q, Boylan NJ, Suk JS, Wang YY, Nance EA, Yang JC, McDonnell PJ, Cone RA, Duh EJ, Hanes J. Nanoparticle diffusion in, and microrheology of, the bovine vitreous ex vivo. J Control Release 2013. [PMID: 23369761 DOI: 10.1016/jjconrel.2013.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Intravitreal injection of biodegradable nanoparticles (NP) holds promise for gene therapy and drug delivery to the back of the eye. In some cases, including gene therapy, NP need to diffuse rapidly from the site of injection in order to reach targeted cell types in the back of the eye, whereas in other cases it may be preferred for the particles to remain at the injection site and slowly release drugs that may then diffuse to the site of action. We studied the movements of polystyrene (PS) NP of various sizes and surface chemistries in fresh bovine vitreous. PS NP as large as 510nm rapidly penetrated the vitreous gel when coated with polyethylene glycol (PEG), whereas the movements of NP 1190nm in diameter or larger were highly restricted regardless of surface chemistry owing to steric obstruction. PS NP coated with primary amine groups (NH2) possessed positively charged surfaces at the pH of bovine vitreous (pH=7.2), and were immobilized within the vitreous gel. In comparison, PS NP coated with COOH (possessing negatively charged surfaces) in the size range of 100-200nm and at particle concentrations below 0.0025% (w/v) readily diffused through the vitreous meshwork; at higher concentrations (~0.1% w/v), these nanoparticles aggregated within vitreous. Based on the mobility of different sized PEGylated PS NP (PS-PEG), we estimated the average mesh size of fresh bovine vitreous to be ~550±50nm. The bovine vitreous behaved as an impermeable elastic barrier to objects sized 1190nm and larger, but as a highly permeable viscoelastic liquid to non-adhesive objects smaller than 510nm in diameter. Guided by these studies, we next sought to examine the transport of drug- and DNA-loaded nanoparticles in bovine vitreous. Biodegradable NP with a diameter of 227nm, composed of a poly(lactic-co-glycolic acid) (PLGA)-based core coated with poly(vinyl alcohol) rapidly penetrated vitreous. Rod-shaped, highly-compacted CK30PEG10k/DNA with PEG coating (neutral surface charge; hydrodynamic diameter ~60nm) also diffused rapidly within vitreous. These findings will help guide the development of nanoparticle-based therapeutics for the treatment of vision-threatening ocular diseases.
Collapse
Affiliation(s)
- Qingguo Xu
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chennamaneni SR, Mamalis C, Archer B, Oakey Z, Ambati BK. Development of a novel bioerodible dexamethasone implant for uveitis and postoperative cataract inflammation. J Control Release 2013; 167:53-9. [DOI: 10.1016/j.jconrel.2013.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/30/2012] [Accepted: 01/05/2013] [Indexed: 11/29/2022]
|
42
|
Xu Q, Boylan NJ, Suk JS, Wang YY, Nance EA, Yang JC, McDonnell PJ, Cone RA, Duh EJ, Hanes J. Nanoparticle diffusion in, and microrheology of, the bovine vitreous ex vivo. J Control Release 2013; 167:76-84. [PMID: 23369761 DOI: 10.1016/j.jconrel.2013.01.018] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/21/2012] [Accepted: 01/20/2013] [Indexed: 01/28/2023]
Abstract
Intravitreal injection of biodegradable nanoparticles (NP) holds promise for gene therapy and drug delivery to the back of the eye. In some cases, including gene therapy, NP need to diffuse rapidly from the site of injection in order to reach targeted cell types in the back of the eye, whereas in other cases it may be preferred for the particles to remain at the injection site and slowly release drugs that may then diffuse to the site of action. We studied the movements of polystyrene (PS) NP of various sizes and surface chemistries in fresh bovine vitreous. PS NP as large as 510nm rapidly penetrated the vitreous gel when coated with polyethylene glycol (PEG), whereas the movements of NP 1190nm in diameter or larger were highly restricted regardless of surface chemistry owing to steric obstruction. PS NP coated with primary amine groups (NH2) possessed positively charged surfaces at the pH of bovine vitreous (pH=7.2), and were immobilized within the vitreous gel. In comparison, PS NP coated with COOH (possessing negatively charged surfaces) in the size range of 100-200nm and at particle concentrations below 0.0025% (w/v) readily diffused through the vitreous meshwork; at higher concentrations (~0.1% w/v), these nanoparticles aggregated within vitreous. Based on the mobility of different sized PEGylated PS NP (PS-PEG), we estimated the average mesh size of fresh bovine vitreous to be ~550±50nm. The bovine vitreous behaved as an impermeable elastic barrier to objects sized 1190nm and larger, but as a highly permeable viscoelastic liquid to non-adhesive objects smaller than 510nm in diameter. Guided by these studies, we next sought to examine the transport of drug- and DNA-loaded nanoparticles in bovine vitreous. Biodegradable NP with a diameter of 227nm, composed of a poly(lactic-co-glycolic acid) (PLGA)-based core coated with poly(vinyl alcohol) rapidly penetrated vitreous. Rod-shaped, highly-compacted CK30PEG10k/DNA with PEG coating (neutral surface charge; hydrodynamic diameter ~60nm) also diffused rapidly within vitreous. These findings will help guide the development of nanoparticle-based therapeutics for the treatment of vision-threatening ocular diseases.
Collapse
Affiliation(s)
- Qingguo Xu
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kulkarni SS, Kompella UB. Nanoparticles for Drug and Gene Delivery in Treating Diseases of the Eye. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2013. [DOI: 10.1007/7653_2013_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
44
|
Yandrapu S, Kompella UB. Development of sustained-release microspheres for the delivery of SAR 1118, an LFA-1 antagonist intended for the treatment of vascular complications of the eye. J Ocul Pharmacol Ther 2012; 29:236-48. [PMID: 23256487 DOI: 10.1089/jop.2012.0210] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The objective of this study was to design 1, 3, and 6 month sustained-release poly (lactide-co-glycolide) (PLGA) microspheres of SAR 1118, a lymphocyte function-associated antigen-1 antagonist, using Design of Experiments. A full-factorial design was used to identify the polymers suitable for degradation in 1, 3, and 6 months and the Box-Behnken design was used to study the influence of the polymer type, polymer concentration, and drug to polymer ratio on drug loading, burst release, and particle size. From the full-factorial design, PLGA (50:50), PLGA (75:25), and PLGA (85:15) with an inherent viscosity of 0.3-0.5 dL/g were identified as polymers suitable for degradation in 1, 3, and 6 months, respectively. From the Box-Behnken design, the optimized polymer concentration (12% w/v) and drug to polymer ratio (0.15) were identified and used to prepare the SAR 1118-encapsulated microspheres with the above 3 polymers and evaluated for drug loading, burst release, and sustained drug release. The burst release in these 3 batches was less than 20% and the drug loading ranged from 15%-18%. More than 90% of SAR 1118 release from PLGA (50:50), PLGA (75:25), and PLGA (85:15) microspheres occurred in 1, 3, and 6 months, respectively. Thus, the in vitro cumulative release data are remarkably close to the predicted values. The results demonstrated the potential of the Design of Experiments in designing the SAR 1118 microspheres with a high loading efficiency, low burst release, and sustained release for a desired duration.
Collapse
Affiliation(s)
- Sarath Yandrapu
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | |
Collapse
|
45
|
Tyagi P, Kadam RS, Kompella UB. Comparison of suprachoroidal drug delivery with subconjunctival and intravitreal routes using noninvasive fluorophotometry. PLoS One 2012; 7:e48188. [PMID: 23118950 PMCID: PMC3485142 DOI: 10.1371/journal.pone.0048188] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 09/26/2012] [Indexed: 01/08/2023] Open
Abstract
Purpose To determine whether exposure of sodium fluorescein (NaF) to the choroid-retina region in the posterior segment of the eye is greater with suprachoroidal injection when compared to intravitreal and transscleral routes. Methods Suprachoroidal injection, a new approach for drug delivery to the posterior segment of the eye was validated using a 34 G needle and Indian ink injections in Sprague Dawley rats, followed by histology. Delivery of NaF was compared in Sprague Dawley rats after suprachoroidal, posterior subconjunctival, or intravitreal injections. NaF levels were monitored noninvasively up to 6 hours using Fluorotron Master™, an ocular fluorophotometer Pharmacokinetic parameters were estimated using WinNonlin. Results Histological analysis indicated localization of India ink to the suprachoroidal space below sclera, following injection. NaF delivery to choroid-retina was in the order: suprachoroidal > intravitreal >posterior subconjunctival injection. Peak NaF concentration (Cmax) in choroid-retina was 36-fold (p = 0.001) and 25-fold (p = 0.001) higher after suprachoroidal (2744±1111 ng/ml) injection when compared to posterior subconjunctival (76±6 ng/ml) and intravitreal (108±39 ng/ml) injections, respectively. NaF exposure (AUC0–360min) to choroid-retina after suprachoroidal injection was 6-fold (p = 0.001) and 2-fold (p = 0.03) higher than posterior subconjunctival and intravitreal injections, respectively. Choroid-retina Tmax was observed immediately after dosing with suprachoroidal injections and at 10 and 27.5 minutes, respectively, with subconjunctival and intravitreal injections. Conclusions Suprachoroidal injections are feasible in a rat model. Suprachoroidal injections resulted in the highest bioavailability, that is, the extent and rate of delivery of NaF to choroid-retina, when compared to intravitreal and posterior subconjunctival injections. Ocular fluorophotometry is useful for noninvasive monitoring of NaF in rats following administration by various routes including suprachoroidal route.
Collapse
Affiliation(s)
- Puneet Tyagi
- Nanomedicine and Drug Delivery Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Rajendra S. Kadam
- Nanomedicine and Drug Delivery Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Uday B. Kompella
- Nanomedicine and Drug Delivery Laboratory, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
46
|
Kadam RS, Tyagi P, Edelhauser HF, Kompella UB. RETRACTED: Influence of choroidal neovascularization and biodegradable polymeric particle size on transscleral sustained delivery of triamcinolone acetonide. Int J Pharm 2012; 434:140-7. [PMID: 22633904 PMCID: PMC3573139 DOI: 10.1016/j.ijpharm.2012.05.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 04/19/2012] [Accepted: 05/15/2012] [Indexed: 11/29/2022]
Abstract
PURPOSE One objective of this study was to determine whether polymeric nanoparticles and/or microparticles sustain transscleral choroidal and retinal delivery of triamcinolone acetonide (TA) for two months in therapeutically effective concentrations after single periocular administration. Another objective of this study was to assess the influence of choroidal neovascularization on transscleral delivery of TA. METHODS Polymeric nano- and micro-particles of TA were prepared by o/w emulsion-solvent evaporation method using poly-l-lactide (PLA). Particles were characterized for drug loading, size, surface morphology, and the in vitro drug release profile. Choroidal neovascularization (CNV) was induced in brown Norway (BN) rats using a 532 nm diode argon laser and the CNV induction was assessed using fluorescein angiography. In vivo delivery was assessed in control and CNV induced rats at 2 months after periocular injection of TA loaded nano- or micro-particle suspension, or plain TA suspension in PBS (pH 7.4). Ocular tissue levels of TA were estimated using LC-MS/MS following liquid-liquid extraction of drug from tissue samples. Nile red loaded microparticles entrapped in periocular tissue at the end of the study was visualized using scanning electron microscopy and confocal microscopy. Inhibitory effect of TA on VEGF secretion was evaluated in ARPE-19 cells. RESULTS Triamcinolone acetonide-PLA nano- (551 nm) and micro-particles (2090 nm), with 14.7 and 29.5% drug loading, respectively, sustained in vitro TA release for about 45 and 120 days. After subconjunctival injection, microparticles were able to sustain the delivery in all intraocular tissues for 2 months; whereas no drug levels were detected for TA loaded nanoparticles and plain suspension of TA. Intraocular delivery of TA from microparticles was higher in CNV induced rats when compared to control rats. Significant amount of microparticles remained in periocular tissue at 2 months after injection, and maintained spherical shape. TA decreased VEGF secretion by 50% at 0.07 μM. At the end of the in vivo study, choroid-RPE and retina TA levels in CNV induced rats were 16- and 5-fold higher than the IC(50) for VEGF secretion. CONCLUSIONS Single periocular injection of polymeric microparticles but not nanoparticles sustained effective levels of TA in choroid-RPE and retina for 2 months, with the TA delivery being greater in CNV induced rats than the control rats.
Collapse
Affiliation(s)
- Rajendra S. Kadam
- Departments of Pharmaceutical Sciences and Ophthalmology University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Puneet Tyagi
- Departments of Pharmaceutical Sciences and Ophthalmology University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Uday B. Kompella
- Departments of Pharmaceutical Sciences and Ophthalmology University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|