1
|
Dolatabadi EN, Akbarzadeh Zaky MR, Abbas FH, Milani AE, André H, Alizadeh E. Recent Advances on Modeling Retinal Disease: Towards Efficient Gene/Drug Therapy. Exp Eye Res 2025; 256:110416. [PMID: 40320033 DOI: 10.1016/j.exer.2025.110416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/22/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Advanced modeling biotechnologies are required to understand retinal diseases and develop effective treatments based on the patient's genetic background, lifestyle, and environment. In this work, recent advances in different types of study models that are used in the retinal disease area of research will be explored. The retinal models to be covered are: in vivo systems (human and animal), in vitro organisms (cell lines, primary cells, patient-derived stem cells, microfluidics, organoids, and spheroids), ex vivo models (explant cultures and retinal tissue preparations), and in silico models (computational and mathematical). Moreover, the unique comprehension of models of retinal disease, advantages, and disadvantages will be scrutinized. Finally, innovations/improvements derived from models towards gene and pharmacological therapy that display promise for treating retinal illnesses are elucidated.
Collapse
Affiliation(s)
- Elham Norouz Dolatabadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fatima Hashim Abbas
- Department of Aesthetic and Laser Techniques, College of Health and Medical Techniques, Al-Mustagbal University, Babylon, Iraq
| | | | - Helder André
- Department of Clinical Neuroscience, Karolinska Institute, Karolinska, Sweden
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Piroozmand S, Soheili ZS, Latifi-Navid H, Samiei S, Rezaei-Kanavi M, Behrooz AB, Hosseinkhani S. MiRGD peptideticle targeted delivery of hinge-truncated soluble VEGF receptor 1 fusion protein to the retinal pigment epithelium cell line and newborn mice retina. Int J Biol Macromol 2025; 307:141916. [PMID: 40068751 DOI: 10.1016/j.ijbiomac.2025.141916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
Eye-related Angiogenesis and vascular permeability changes lead to retinal vascular disorders. There is an important need to design a novel targeted anti-VEGF drug delivery system to inhibit neovascularization in the retina. The peptide-based carriers are promising for gene therapy due to their flexibility in design, ease of production, structural diversity, low toxicity, and immunogenicity. The hinge-truncated soluble VEGF receptor 1 (htsFLT01) protein, has the ability to bind to both VEGF and PlGF molecules. In the present study, htsFLT01 gene delivery by targeted MiRGD peptide carrier was investigated in the mouse Retinal Pigment Epithelium (mRPE) cell line and mouse model to evaluate the potential of the newly developed peptideticle as an effective therapeutic platform for gene delivery. The characterization results demonstrated that the peptide carrier condensed htsFLT01 DNA, neutralizes its negative charge, and protected it from endonucleases. The size and charge of the nanocomplexes were optimized to effectively target the retina. Based on tube formation assay, migration analyses and intravitreal injection of MiRGD-htsFLT01 nanocomplex into the newborn mice eye, the function of htsFLT01 was investigated. The reduction of tube-like structures in HUVEC cells was notably observed following VEGF neutralization and the findings demonstrated an association between the expression of htsFLT01 and the inhibition of RPE cell migration. The vascular development was inhibited in the deep, intermediate, and superficial capillary plexus layers in the retina. The novel drug MiRGD/htsFLT01 complex, represents a promising potential platform for targeted gene therapy in the eye due to its biocompatibility, likely safety and highly effective function.
Collapse
Affiliation(s)
- Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zahra-Soheila Soheili
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran 1416634793, Iran; School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran 1953833511, Iran
| | - Shahram Samiei
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mozhgan Rezaei-Kanavi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Barzegar Behrooz
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Khaparde A, Mathias GP, Poornachandra B, Thirumalesh MB, Shetty R, Ghosh A. Gene therapy for retinal diseases: From genetics to treatment. Indian J Ophthalmol 2024; 72:1091-1101. [PMID: 39078952 PMCID: PMC11451791 DOI: 10.4103/ijo.ijo_2902_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 04/19/2024] [Indexed: 10/06/2024] Open
Abstract
The gene therapy approach for retinal disorders has been considered largely over the last decade owing to the favorable outcomes of the US Food and Drug Administration-approved commercial gene therapy, Luxturna. Technological advances in recent years, such as next-generation sequencing, research in molecular pathogenesis of retinal disorders, and precise correlations with their clinical phenotypes, have contributed to the progress of gene therapies for various diseases worldwide, and more recently in India as well. Thus, considerable research is being conducted for the right choice of vectors, transgene engineering, and accessible and cost-effective large-scale vector production. Many retinal disease-specific clinical trials are presently being conducted, thereby necessitating the collation of such information as a ready reference for the scientific and clinical community. In this article, we present an overview of existing gene therapy research, which is derived from an extensive search across PubMed, Google Scholar, and clinicaltrials.gov sources. This contributes to prime the understanding of basic aspects of this cutting-edge technology and information regarding current clinical trials across many different conditions. This information will provide a comprehensive evaluation of therapies in existing use/research for personalized treatment approaches in retinal disorders.
Collapse
Affiliation(s)
- Ashish Khaparde
- GROW Research Laboratory, Narayana Nethralaya Foundation, Manipal, Karnataka, India
| | - Grace P Mathias
- GROW Research Laboratory, Narayana Nethralaya Foundation, Manipal, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - B Poornachandra
- Department of Vitreo Retina Services, Narayana Nethralaya, Manipal, Karnataka, India
| | - M B Thirumalesh
- Department of Vitreo Retina Services, Narayana Nethralaya, Manipal, Karnataka, India
| | - Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya, Bengaluru, Karnataka, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Manipal, Karnataka, India
| |
Collapse
|
4
|
Kumar B, Mishra M, Talreja D, Cashman S, Kumar-Singh R. Cell-Penetrating Chaperone Nuc1 for Small- and Large-Molecule Delivery Into Retinal Cells and Tissues. Invest Ophthalmol Vis Sci 2024; 65:31. [PMID: 39028980 PMCID: PMC11262537 DOI: 10.1167/iovs.65.8.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/20/2024] [Indexed: 07/21/2024] Open
Abstract
Purpose There are currently no means available for the efficient delivery of recombinant proteins into retinal cells in vivo. Although cell-penetrating peptides have been somewhat effective in protein delivery to the retina, they generally require conjugation chemistry with the payload, negatively impacting function of the therapeutic protein. In this study, we developed a novel peptide (Nuc1) that acts as a chaperone for delivery of small and large molecules, including steroids, peptides, antibodies, recombinant proteins, and viruses (adeno-associated viruses [AAVs]) across biological membranes in vivo without the need for conjugation. Methods Nuc1 peptide was designed based on sequences known to bind heparan sulfate proteoglycans and nucleolin found on the surface of retinal cells. Nuc1 was injected into the vitreous of mice with a variety of molecules and retinas examined for uptake and function of these molecules. Results Nuc1 engages the process of macropynocytosis for cell entry. The delivery of functional recombinant X-linked inhibitor of apoptosis protein to photoreceptors via the intravitreal route of injection inhibited retinal apoptosis. Nuc1 was found to enhance the delivery of anti-VEGF antibodies delivered intravitreally or topically in models of age-related macular degeneration (AMD). Nuc1 enhanced delivery of decorin, facilitating significant inhibition of neovascularization and fibrosis in a model of AMD. Finally, Nuc1 was found to enhance penetration of retinal cells and tissues by AAV via both the subretinal and intravitreal routes of injection. Conclusions Nuc1 shows promise as a novel approach for the delivery of recombinant proteins into retinal cells in vivo.
Collapse
Affiliation(s)
- Binit Kumar
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Manish Mishra
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Deepa Talreja
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Siobhan Cashman
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Rajendra Kumar-Singh
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
5
|
Rohira H, Shankar S, Yadav S, Srivastava PP, Minocha S, Vaddavalli PK, Shah SG, Chugh A. RiTe conjugate mediated corneal collagen crosslinking, a novel therapeutic intervention for keratoconus - in vitro and in vivo study. Int J Pharm 2024; 656:124092. [PMID: 38583820 DOI: 10.1016/j.ijpharm.2024.124092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Corneal collagen crosslinking (CXL) is an effective method to halt the disease progression of keratoconus, a progressive corneal dystrophy leading to cone shaped cornea. Despite the efficacy of standard protocol, the concerning step of this procedure is epithelial debridement performed to facilitate the entry of riboflavin drug. Riboflavin, a key molecule in CXL protocol, is a sparsely permeable hydrophilic drug in corneal tissues. The present study has employed cell penetrating peptide (CPP), Tat2, to enhance the penetration of riboflavin molecule, and thereby improve currently followed CXL protocol. This study demonstrates approximately two-fold enhanced uptake of CPP riboflavin conjugate, Tat2riboflavin-5'Phosphate (RiTe conjugate), both in vitro and in vivo. Two different CXL protocols (Epi ON and Epi OFF) have been introduced and implemented in rabbit corneas using RiTe conjugate in the present study. The standard and RiTe conjugate mediated CXL procedures exhibited an equivalent extent of crosslinking in both the methods. Reduced keratocyte loss and no endothelial damage in RiTe conjugate mediated CXL further ascertains the safety of the proposed CXL protocols. Therefore, RiTe conjugate mediated CXL protocols present as potential alternatives to the standard keratoconus treatment in providing equally effective, less invasive and patient compliant treatment modality.
Collapse
Affiliation(s)
- Harsha Rohira
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sujithra Shankar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Shikha Yadav
- National Institute of Biologicals, NOIDA, Uttar Pradesh 201309, India
| | - Priyanka P Srivastava
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Shilpi Minocha
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India
| | | | - Sushmita G Shah
- Dr C M Shah Memorial Charitable Trust - Netra Mandir, Madona Colony Road, Borivali West, Mumbai, Maharashtra 400092, India.
| | - Archana Chugh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
6
|
Chen Y, Ye Z, Chen H, Li Z. Breaking Barriers: Nanomedicine-Based Drug Delivery for Cataract Treatment. Int J Nanomedicine 2024; 19:4021-4040. [PMID: 38736657 PMCID: PMC11086653 DOI: 10.2147/ijn.s463679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/27/2024] [Indexed: 05/14/2024] Open
Abstract
Cataract is a leading cause of blindness globally, and its surgical treatment poses a significant burden on global healthcare. Pharmacologic therapies, including antioxidants and protein aggregation reversal agents, have attracted great attention in the treatment of cataracts in recent years. Due to the anatomical and physiological barriers of the eye, the effectiveness of traditional eye drops for delivering drugs topically to the lens is hindered. The advancements in nanomedicine present novel and promising strategies for addressing challenges in drug delivery to the lens, including the development of nanoparticle formulations that can improve drug penetration into the anterior segment and enable sustained release of medications. This review introduces various cutting-edge drug delivery systems for cataract treatment, highlighting their physicochemical properties and surface engineering for optimal design, thus providing impetus for further innovative research and potential clinical applications of anti-cataract drugs.
Collapse
Affiliation(s)
- Yilin Chen
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
- Senior Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zi Ye
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
- Senior Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Haixu Chen
- Institute of Geriatrics, National Clinical Research Center for Geriatrics Diseases, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zhaohui Li
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
- Senior Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
7
|
Vishwakarma M, Agrawal P, Soni S, Tomar S, Haider T, Kashaw SK, Soni V. Cationic nanocarriers: A potential approach for targeting negatively charged cancer cell. Adv Colloid Interface Sci 2024; 327:103160. [PMID: 38663154 DOI: 10.1016/j.cis.2024.103160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024]
Abstract
Cancer, a widespread and lethal disease, necessitates precise therapeutic interventions to mitigate its devastating impact. While conventional chemotherapy remains a cornerstone of cancer treatment, its lack of specificity towards cancer cells results in collateral damage to healthy tissues, leading to adverse effects. Thus, the quest for targeted strategies has emerged as a critical focus in cancer research. This review explores the development of innovative targeting methods utilizing novel drug delivery systems tailored to recognize and effectively engage cancer cells. Cancer cells exhibit morphological and metabolic traits, including irregular morphology, unchecked proliferation, metabolic shifts, genetic instability, and a higher negative charge, which serve as effective targeting cues. Central to these strategies is the exploitation of the unique negative charge characteristic of cancer cells, attributed to alterations in phospholipid composition and the Warburg effect. Leveraging this distinct feature, researchers have devised cationic carrier systems capable of enhancing the specificity of therapeutic agents towards cancer cells. The review delineates the underlying causes of the negative charge in cancer cells and elucidates various targeting approaches employing cationic compounds for drug delivery systems. Furthermore, it delves into the methods employed for the preparation of these systems. Beyond cancer treatment, the review also underscores the multifaceted applications of cationic carrier systems, encompassing protein and peptide delivery, imaging, photodynamic therapy, gene delivery, and antimicrobial applications. This comprehensive exploration underscores the potential of cationic carrier systems as versatile tools in the fight against cancer and beyond.
Collapse
Affiliation(s)
- Monika Vishwakarma
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Poornima Agrawal
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Sakshi Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Surbhi Tomar
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Tanweer Haider
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India; Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior 474005, MP, India
| | - Sushil K Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, MP, India.
| |
Collapse
|
8
|
Toffoletto N, Salema-Oom M, Nicoli S, Pescina S, González-Fernández FM, Pinto CA, Saraiva JA, Alves de Matos AP, Vivero-Lopez M, Huete-Toral F, Carracedo G, Saramago B, Serro AP. Dexamethasone phosphate and penetratin co-eluting contact lenses: a strategy to enhance ocular drug permeability. Int J Pharm 2024; 650:123685. [PMID: 38072146 DOI: 10.1016/j.ijpharm.2023.123685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/29/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023]
Abstract
Contact lenses (CLs) have been suggested as drug delivery platforms capable of increasing the drug residence time on the cornea and therefore its bioavailability. However, when targeting the posterior segment of the eye, the drug released from CLs still encounters the barrier effect of the ocular tissues, which considerably reduces the efficacy of administration. This work aims at the development of CLs able to simultaneously deliver an anti-inflammatory drug (dexamethasone sodium phosphate) and a cell-penetrating peptide (penetratin), the latter acting as a drug carrier across the tissues. Hydroxyethyl methacrylate (HEMA)-based hydrogels were functionalized with acrylic acid (AAc) and/or aminopropyl methacrylamide (APMA) to serve as CL materials with increased affinity for the drug and peptide. APMA-functionalized hydrogels sustained the dual release for 8 h, which is compatible with the wearing time of daily CLs. Hydrogels demonstrated suitable light transmittance, swelling capacity and in vitro biocompatibility. The anti-inflammatory activity of the drug was not compromised by the presence of the peptide nor by sterilization. The ocular distribution of the drug after 6 h of CL wearing was evaluated in vivo in rabbits and revealed that the amount of drug in the cornea and aqueous humor significantly increased when the drug was co-delivered with penetratin.
Collapse
Affiliation(s)
- Nadia Toffoletto
- Centro de Química Estrutural, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health & Science, Campus Universitario, 2829-511 Caparica, Portugal.
| | - Madalena Salema-Oom
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health & Science, Campus Universitario, 2829-511 Caparica, Portugal.
| | - Sara Nicoli
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/a, 43124 Parma, Italy.
| | - Silvia Pescina
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/a, 43124 Parma, Italy.
| | - Felipe M González-Fernández
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/a, 43124 Parma, Italy.
| | - Carlos A Pinto
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Jorge A Saraiva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António P Alves de Matos
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health & Science, Campus Universitario, 2829-511 Caparica, Portugal.
| | - Maria Vivero-Lopez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Insititute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Fernando Huete-Toral
- Ocupharm Research Group, Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Complutense University of Madrid, C/Arcos de Jalón 118, 28037 Madrid, Spain.
| | - Gonzalo Carracedo
- Ocupharm Research Group, Department of Optometry and Vision, Faculty of Optics and Optometry, Complutense University of Madrid, C/Arcos de Jalón 118, 28037 Madrid, Spain.
| | - Benilde Saramago
- Centro de Química Estrutural, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.
| | - Ana Paula Serro
- Centro de Química Estrutural, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz School of Health & Science, Campus Universitario, 2829-511 Caparica, Portugal.
| |
Collapse
|
9
|
Zhou J, Cai Y, Li T, Zhou H, Dong H, Wu X, Li Z, Wang W, Yuan D, Li Y, Shi J. Aflibercept Loaded Eye-Drop Hydrogel Mediated with Cell-Penetrating Peptide for Corneal Neovascularization Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302765. [PMID: 37679056 DOI: 10.1002/smll.202302765] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Corneal neovascularization (CoNV) is a major cause of visual impairment worldwide. Currently, available treatment options have limited efficacy and are associated with adverse effects due to biological barriers and clearance mechanisms. To address this challenge, a novel topical delivery system is developed-Gel 2_1&Eylea-an aflibercept-loaded eye-drop hydrogel mediated with cell-penetrating peptide 1. Gel 2_1&Eylea demonstrates superior membrane permeability, increased stability, and prolonged drug retention time on the ocular surface, and thus may improve drug efficacy. In a rabbit CoNV model, Gel 2_1&Eylea significantly reduces the density of neovascularization with no adverse effects on normal corneoscleral limbal vessels, demonstrating high efficacy and biocompatibility. This work identifies a promising treatment for CoNV which has the potential to benefit other ocular neovascular diseases.
Collapse
Affiliation(s)
- Jianan Zhou
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Yuting Cai
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Tingting Li
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Haixiang Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Huilei Dong
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Xia Wu
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
- Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, Guangdong, 518116, China
| | - Zenghui Li
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Wenjie Wang
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Dan Yuan
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Yun Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Junfeng Shi
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| |
Collapse
|
10
|
Boddu SH, Acharya D, Hala V, Jani H, Pande S, Patel C, Shahwan M, Jwala R, Ranch KM. An Update on Strategies to Deliver Protein and Peptide Drugs to the Eye. ACS OMEGA 2023; 8:35470-35498. [PMID: 37810716 PMCID: PMC10552503 DOI: 10.1021/acsomega.3c02897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023]
Abstract
In the past few decades, advancements in protein engineering, biotechnology, and structural biochemistry have resulted in the discovery of various techniques that enhanced the production yield of proteins, targetability, circulating half-life, product purity, and functionality of proteins and peptides. As a result, the utilization of proteins and peptides has increased in the treatment of many conditions, including ocular diseases. Ocular delivery of large molecules poses several challenges due to their high molecular weight, hydrophilicity, unstable nature, and poor permeation through cellular and enzymatic barriers. The use of novel strategies for delivering protein and peptides such as glycoengineering, PEGylation, Fc-fusion, chitosan nanoparticles, and liposomes have improved the efficacy, safety, and stability, which consequently expanded the therapeutic potential of proteins. This review article highlights various proteins and peptides that are useful in ocular disorders, challenges in their delivery to the eye, and strategies to enhance ocular bioavailability using novel delivery approaches. In addition, a few futuristic approaches that will assist in the ocular delivery of proteins and peptides were also discussed.
Collapse
Affiliation(s)
- Sai H.
S. Boddu
- College
of Pharmacy and Health Sciences, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Center
of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Devarshi Acharya
- Department
of Pharmaceutics, L. M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Vivek Hala
- Department
of Pharmaceutics, L. M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Harshil Jani
- Department
of Pharmaceutics, L. M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
- Gujarat
Technological University, Ahmedabad, Gujarat 382424, India
| | - Sonal Pande
- Gujarat
Technological University, Ahmedabad, Gujarat 382424, India
- Department
of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Chirag Patel
- Department
of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Moyad Shahwan
- College
of Pharmacy and Health Sciences, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Center
of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Renukuntla Jwala
- School
of
Pharmacy, The University of Texas at El
Paso, 1101 N Campbell
St., El Paso, Texas 79902, United States
- Department
of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, High Point, North Carolina, 27240, United States
| | - Ketan M. Ranch
- Department
of Pharmaceutics, L. M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| |
Collapse
|
11
|
Lee H, Noh H. Advancements in Nanogels for Enhanced Ocular Drug Delivery: Cutting-Edge Strategies to Overcome Eye Barriers. Gels 2023; 9:718. [PMID: 37754399 PMCID: PMC10529109 DOI: 10.3390/gels9090718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 08/29/2023] [Accepted: 09/02/2023] [Indexed: 09/28/2023] Open
Abstract
Nanomedicine in gel or particle formation holds considerable potential for enhancing passive and active targeting within ocular drug delivery systems. The complex barriers of the eye, exemplified by the intricate network of closely connected tissue structures, pose significant challenges for drug administration. Leveraging the capability of engineered nanomedicine offers a promising approach to enhance drug penetration, particularly through active targeting agents such as protein peptides and aptamers, which facilitate targeted release and heightened bioavailability. Simultaneously, DNA carriers have emerged as a cutting-edge class of active-targeting structures, connecting active targeting agents and illustrating their potential in ocular drug delivery applications. This review aims to consolidate recent findings regarding the optimization of various nanoparticles, i.e., hydrogel-based systems, incorporating both passive and active targeting agents for ocular drug delivery, thereby identifying novel mechanisms and strategies. Furthermore, the review delves into the potential application of DNA nanostructures, exploring their role in the development of targeted drug delivery approaches within the field of ocular therapy.
Collapse
Affiliation(s)
| | - Hyeran Noh
- Department of Optometry, Seoul National University of Science and Technology, Gongnung-ro 232, Nowon-gu, Seoul 01811, Republic of Korea;
| |
Collapse
|
12
|
Asrorov AM, Wang H, Zhang M, Wang Y, He Y, Sharipov M, Yili A, Huang Y. Cell penetrating peptides: Highlighting points in cancer therapy. Drug Dev Res 2023; 84:1037-1071. [PMID: 37195405 DOI: 10.1002/ddr.22076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/29/2023] [Indexed: 05/18/2023]
Abstract
Cell-penetrating peptides (CPPs), first identified in HIV a few decades ago, deserved great attention in the last two decades; especially to support the penetration of anticancer drug means. In the drug delivery discipline, they have been involved in various approaches from mixing with hydrophobic drugs to the use of genetically conjugated proteins. The early classification as cationic and amphipathic CPPs has been extended to a few more classes such as hydrophobic and cyclic CPPs so far. Developing potential sequences utilized almost all methods of modern science: choosing high-efficiency peptides from natural protein sequences, sequence-based comparison, amino acid substitution, obtaining chemical and/or genetic conjugations, in silico approaches, in vitro analysis, animal experiments, etc. The bottleneck effect in this discipline reveals the complications that modern science faces in drug delivery research. Most CPP-based drug delivery systems (DDSs) efficiently inhibited tumor volume and weight in mice, but only in rare cases reduced their levels and continued further processes. The integration of chemical synthesis into the development of CPPs made a significant contribution and even reached the clinical stage as a diagnostic tool. But constrained efforts still face serious problems in overcoming biobarriers to reach further achievements. In this work, we reviewed the roles of CPPs in anticancer drug delivery, focusing on their amino acid composition and sequences. As the most suitable point, we relied on significant changes in tumor volume in mice resulting from CPPs. We provide a review of individual CPPs and/or their derivatives in a separate subsection.
Collapse
Affiliation(s)
- Akmal M Asrorov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Bioorganic Chemistry, AS of Uzbekistan, Tashkent, Uzbekistan
- Department of Natural Substances Chemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Huiyuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Meng Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yonghui Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yang He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mirkomil Sharipov
- Institute of Bioorganic Chemistry, AS of Uzbekistan, Tashkent, Uzbekistan
| | - Abulimiti Yili
- The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, Xinjiang, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Zhongshan Institute for Drug Discovery, Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai, China
| |
Collapse
|
13
|
Fan X, Jiang K, Geng F, Lu W, Wei G. Ocular therapies with biomacromolecules: From local injection to eyedrop and emerging noninvasive delivery strategies. Adv Drug Deliv Rev 2023; 197:114864. [PMID: 37156266 DOI: 10.1016/j.addr.2023.114864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/15/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
The last two decades have witnessed a continuously increasing number of biomacromolecules approved for the treatment of ocular diseases. The eye possesses multiple protective mechanisms to resist the invasion of exogenous substances, but meanwhile these physiological defense systems also act as strong barriers, impeding absorption of most biomacromolecules into the eye. As a result, local injections play predominant roles for posterior ocular delivery of biomacromolecules in clinical practice. To achieve safe and convenient application of biomacromolecules, alternative strategies to realize noninvasive intraocular delivery are necessary. Various nanocarriers, novel penetration enhancers and physical strategies have been explored to facilitate delivery of biomacromolecules to both anterior and posterior ocular segments but still suffered difficulties in clinical translation. This review compares the anatomical and physiological characteristics of the eyes from those frequently adopted experimental species and profiles the well-established animal models of ocular diseases. We also summarize the ophthalmic biomacromolecules launched on the market and put emphasis on emerging noninvasive intraocular delivery strategies of peptides, proteins and genes.
Collapse
Affiliation(s)
- Xingyan Fan
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Kuan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China; Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200030, P.R. China
| | - Feiyang Geng
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China; The Institutes of Integrative Medicine of Fudan University, Shanghai, 200040, PR China
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, PR China; The Institutes of Integrative Medicine of Fudan University, Shanghai, 200040, PR China; Shanghai Engineering Research Center of ImmunoTherapeutics, Shanghai, 201203, PR China.
| |
Collapse
|
14
|
Hsueh HT, Chou RT, Rai U, Liyanage W, Kim YC, Appell MB, Pejavar J, Leo KT, Davison C, Kolodziejski P, Mozzer A, Kwon H, Sista M, Anders NM, Hemingway A, Rompicharla SVK, Edwards M, Pitha I, Hanes J, Cummings MP, Ensign LM. Machine learning-driven multifunctional peptide engineering for sustained ocular drug delivery. Nat Commun 2023; 14:2509. [PMID: 37130851 PMCID: PMC10154330 DOI: 10.1038/s41467-023-38056-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/12/2023] [Indexed: 05/04/2023] Open
Abstract
Sustained drug delivery strategies have many potential benefits for treating a range of diseases, particularly chronic diseases that require treatment for years. For many chronic ocular diseases, patient adherence to eye drop dosing regimens and the need for frequent intraocular injections are significant barriers to effective disease management. Here, we utilize peptide engineering to impart melanin binding properties to peptide-drug conjugates to act as a sustained-release depot in the eye. We develop a super learning-based methodology to engineer multifunctional peptides that efficiently enter cells, bind to melanin, and have low cytotoxicity. When the lead multifunctional peptide (HR97) is conjugated to brimonidine, an intraocular pressure lowering drug that is prescribed for three times per day topical dosing, intraocular pressure reduction is observed for up to 18 days after a single intracameral injection in rabbits. Further, the cumulative intraocular pressure lowering effect increases ~17-fold compared to free brimonidine injection. Engineered multifunctional peptide-drug conjugates are a promising approach for providing sustained therapeutic delivery in the eye and beyond.
Collapse
Affiliation(s)
- Henry T Hsueh
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Renee Ti Chou
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD, USA
| | - Usha Rai
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wathsala Liyanage
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yoo Chun Kim
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew B Appell
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Jahnavi Pejavar
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Kirby T Leo
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Charlotte Davison
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Patricia Kolodziejski
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ann Mozzer
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - HyeYoung Kwon
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Maanasa Sista
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Nicole M Anders
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Avelina Hemingway
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Sri Vishnu Kiran Rompicharla
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malia Edwards
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ian Pitha
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Michael P Cummings
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD, USA.
| | - Laura M Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
15
|
Nhàn NTT, Maidana DE, Yamada KH. Ocular Delivery of Therapeutic Agents by Cell-Penetrating Peptides. Cells 2023; 12:1071. [PMID: 37048144 PMCID: PMC10093283 DOI: 10.3390/cells12071071] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Cell-penetrating peptides (CPPs) are short peptides with the ability to translocate through the cell membrane to facilitate their cellular uptake. CPPs can be used as drug-delivery systems for molecules that are difficult to uptake. Ocular drug delivery is challenging due to the structural and physiological complexity of the eye. CPPs may be tailored to overcome this challenge, facilitating cellular uptake and delivery to the targeted area. Retinal diseases occur at the posterior pole of the eye; thus, intravitreal injections are needed to deliver drugs at an effective concentration in situ. However, frequent injections have risks of causing vision-threatening complications. Recent investigations have focused on developing long-acting drugs and drug delivery systems to reduce the frequency of injections. In fact, conjugation with CPP could deliver FDA-approved drugs to the back of the eye, as seen by topical application in animal models. This review summarizes recent advances in CPPs, protein/peptide-based drugs for eye diseases, and the use of CPPs for drug delivery based on systematic searches in PubMed and clinical trials. We highlight targeted therapies and explore the potential of CPPs and peptide-based drugs for eye diseases.
Collapse
Affiliation(s)
- Nguyễn Thị Thanh Nhàn
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Daniel E. Maidana
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Kaori H. Yamada
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|
16
|
Tailoring surface properties of liposomes for dexamethasone intraocular administration. J Control Release 2023; 354:323-336. [PMID: 36641118 DOI: 10.1016/j.jconrel.2023.01.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 01/16/2023]
Abstract
Diseases of the posterior eye segment are often characterized by intraocular inflammation, which causes, in the long term, severe impairment of eye functions and, ultimately, vision loss. Aimed at enhancing the delivery of anti-inflammatory drugs to the posterior eye segment upon intravitreal administration, we developed liposomes with an engineered surface to control their diffusivity in the vitreous and retina association. Hydrogenated soybean phosphatidylcholine (HSPC)/cholesterol liposomes were coated with (agmatinyl)6-maltotriosyl-acetamido-N-(octadec-9-en-1-yl)hexanamide (Agm6-M-Oleate), a synthetic non-peptidic cell penetration enhancer (CPE), and/or 5% of mPEG2kDa-DSPE. The zeta potential of liposomes increased, and the mobility in bovine vitreous and colloidal stability decreased with the Agm6-M-Oleate coating concentration. Oppositely, mPEG2kDa-DSPE decreased the zeta potential of liposomes and restored both the diffusivity and the stability in vitreous. Liposomes with 5 mol% Agm6-M-Oleate coating were well tolerated by ARPE-19 retina cells either with or without mPEG2kDa-DSPE, while 10 mol% Agm6-M-Oleate showed cytotoxicity. Agm6-M-Oleate promoted the association of liposomes to ARPE-19 cells with respect to plain liposomes, while mPEG2kDa-DSPE slightly reduced the cell interaction. Dexamethasone hemisuccinate (DH) was remotely loaded into liposomes with a loading capacity of ∼10 wt/wt%. Interestingly, mPEG2kDa-DSPE coating reduced the rate of DH release and enhanced the disposition of Agm6-M-Oleate coated liposomes in the ARPE-19 cell cytosol resulting in a more efficient anti-inflammatory effect. Finally, mPEG2kDa-DSPE enhanced the association of DH-loaded Agm6-M-Oleate coated liposomes to explanted rat retina, which reflected in higher viability of inner and outer nuclear layer cells.
Collapse
|
17
|
Morofuji R, Enomoto H, Honda T, Oyama Y, Ishida R, Kudo K, Okabe K. Exploring Cell-Penetrating Peptides as Penetration Enhancers in Eye Drop Formulations Using a Reconstructed Human Corneal Epithelial Model. Biol Pharm Bull 2023; 46:1720-1730. [PMID: 38044130 DOI: 10.1248/bpb.b23-00457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Ocular tissues function as biological barriers that hinder drug delivery, depending on the target tissue and route of administration, and must be overcome to achieve the desired therapeutic effect. Penetration enhancers have long been investigated to improve corneal drug penetration via eye drop instillation; however, further development is warranted owing to potential safety concerns. In the present study, we focused on cell-penetrating peptides (CPPs) as a penetration enhancer to address the requirements and explored CPP candidates suitable for corneal drug delivery. Using a reconstructed human corneal epithelial tissue model, LabCyte CORNEA-MODEL24 as an alternative to animal testing that is expected to have higher reproducibility than extracted eyeballs and octa-arginine (R8) as a representative model CPP with simple structure, we investigated the enhancement of 6-carboxyfluorescein (6-FAM) uptake by fluorescence imaging and the potential of eye irritation by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Also, surface plasmon resonance (SPR) evaluated the interaction between R8 and model compounds, suggesting that the stronger interaction could facilitate the corneal uptake of compounds. A comparative screening study of corneal uptake using various CPPs showed that the CPPs other than R8 also have the potential to enhance the corneal uptake of 6-FAM. In particular, penetratin (PNT) showed stronger fluorescence intensity. Through these findings, this manuscript provides beneficial information for the development of a novel corneal penetration enhancer with CPPs. In the future, it is expected that the basic findings with R8 will be verified to be applicable to other CPPs for development as penetration enhancers for eye drop formulation.
Collapse
Affiliation(s)
- Ryo Morofuji
- Division of Materials Science, Nara Institute of Science and Technology
- Pharmaceutical Development Division, Nara Research & Development Center, Santen Pharmaceutical Co., Ltd
| | - Hiroshi Enomoto
- Pharmaceutical Development Division, Nara Research & Development Center, Santen Pharmaceutical Co., Ltd
| | - Takahiro Honda
- Division of Materials Science, Nara Institute of Science and Technology
- Pharmaceutical Development Division, Nara Research & Development Center, Santen Pharmaceutical Co., Ltd
| | - Yuki Oyama
- Division of Materials Science, Nara Institute of Science and Technology
| | - Reiji Ishida
- Division of Materials Science, Nara Institute of Science and Technology
| | - Kazuhiro Kudo
- Division of Materials Science, Nara Institute of Science and Technology
- Pharmaceutical Development Division, Nara Research & Development Center, Santen Pharmaceutical Co., Ltd
| | - Komei Okabe
- Division of Materials Science, Nara Institute of Science and Technology
- Pharmaceutical Development Division, Nara Research & Development Center, Santen Pharmaceutical Co., Ltd
| |
Collapse
|
18
|
Wang L, Zhou MB, Zhang H. The Emerging Role of Topical Ocular Drugs to Target the Posterior Eye. Ophthalmol Ther 2021; 10:465-494. [PMID: 34218424 PMCID: PMC8319259 DOI: 10.1007/s40123-021-00365-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of chronic fundus diseases is increasing with the aging of the general population. The treatment of these intraocular diseases relies on invasive drug delivery because of the globular structure and multiple barriers of the eye. Frequent intraocular injections bring heavy burdens to the medical care system and patients. The use of topical drugs to treat retinal diseases has always been an attractive solution. The fast development of new materials and technologies brings the possibility to develop innovative topical formulations. This article reviews anatomical and physiological barriers of the eye which affect the bioavailability of topical drugs. In addition, we summarize innovative topical formulations which enhance the permeability of drugs through the ocular surface and/or extend the drug retention time in the eye. This article also reviews the differences of eyes between different laboratory animals to address the translational challenges of preclinical models. The fast development of in vitro eye models may provide more tools to increase the clinical translationality of topical formulations for intraocular diseases. Clinical successes of topical formulations rely on continuous and collaborative efforts between different disciplines.
Collapse
Affiliation(s)
- Lixiang Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hui Zhang
- Yuanpu Eye Biopharmaceutical Co. Ltd., Chengdu, China.
- , No. 14 Jiuxing Avenue, Gaoxin District, Chengdu, China.
| |
Collapse
|
19
|
Silva M, Peng T, Zhao X, Li S, Farhan M, Zheng W. Recent trends in drug-delivery systems for the treatment of diabetic retinopathy and associated fibrosis. Adv Drug Deliv Rev 2021; 173:439-460. [PMID: 33857553 DOI: 10.1016/j.addr.2021.04.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/05/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Diabetic retinopathy is a frequent microvascular complication of diabetes and a major cause of visual impairment. In advanced stages, the abnormal neovascularization can lead to fibrosis and subsequent tractional retinal detachment and blindness. The low bioavailability of the drugs at the target site imposed by the anatomic and physiologic barriers within the eye, requires long term treatments with frequent injections that often compromise patient's compliance and increase the risk of developing more complications. In recent years, much effort has been put towards the development of new drug delivery platforms aiming to enhance their permeation, to prolong their retention time at the target site and to provide a sustained release with reduced toxicity and improved efficacy. This review provides an overview of the etiology and pathophysiology of diabetic retinopathy and current treatments. It addresses the specific challenges associated to the different ocular delivery routes and provides a critical review of the most recent developments made in the drug delivery field.
Collapse
Affiliation(s)
- Marta Silva
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau
| | - Tangming Peng
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau
| | - Xia Zhao
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau
| | - Shuai Li
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau
| | - Mohd Farhan
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau
| | - Wenhua Zheng
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau.
| |
Collapse
|
20
|
Challenges and strategies for the delivery of biologics to the cornea. J Control Release 2021; 333:560-578. [PMID: 33857565 DOI: 10.1016/j.jconrel.2021.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/05/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023]
Abstract
Biologics, like peptides, proteins and nucleic acids, have proven to be promising drugs for the treatment of numerous diseases. However, besides the off label use of the monoclonal antibody bevacizumab for the treatment of corneal neovascularization, to date no other biologics for corneal diseases have reached the market. Indeed, delivering biologics in the eye remains a challenge, especially at the level of the cornea. While it appears to be a rather accessible tissue for the administration of drugs, the cornea in fact presents several anatomical barriers to delivery. In addition, also intracellular delivery barriers need to be overcome to achieve a promising therapeutic outcome with biologics. This review outlines efforts that have been reported to successfully deliver biologics into the cornea. Biochemical and physical methods for achieving delivery of biologics in the cornea are discussed, with a critical view on their efficacy in overcoming corneal barriers.
Collapse
|
21
|
Peptidomimetics Therapeutics for Retinal Disease. Biomolecules 2021; 11:biom11030339. [PMID: 33668179 PMCID: PMC7995992 DOI: 10.3390/biom11030339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/11/2021] [Accepted: 02/20/2021] [Indexed: 12/28/2022] Open
Abstract
Ocular disorders originating in the retina can result in a partial or total loss of vision, making drug delivery to the retina of vital importance. However, effectively delivering drugs to the retina remains a challenge for ophthalmologists due to various anatomical and physicochemical barriers in the eye. This review introduces diverse administration routes and the accordant pharmacokinetic profiles of ocular drugs to aid in the development of safe and efficient drug delivery systems to the retina with a focus on peptidomimetics as a growing class of retinal drugs, which have great therapeutic potential and a high degree of specificity. We also discuss the pharmacokinetic profiles of small molecule drugs due to their structural similarity to small peptidomimetics. Lastly, various formulation strategies are suggested to overcome pharmacokinetic hurdles such as solubility, retention time, enzymatic degradation, tissue targeting, and membrane permeability. This knowledge can be used to help design ocular delivery platforms for peptidomimetics, not only for the treatment of various retinal diseases, but also for the selection of potential peptidomimetic drug targets.
Collapse
|
22
|
Kantor A, McClements ME, Peddle CF, Fry LE, Salman A, Cehajic-Kapetanovic J, Xue K, MacLaren RE. CRISPR genome engineering for retinal diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 182:29-79. [PMID: 34175046 DOI: 10.1016/bs.pmbts.2021.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Novel gene therapy treatments for inherited retinal diseases have been at the forefront of translational medicine over the past couple of decades. Since the discovery of CRISPR mechanisms and their potential application for the treatment of inherited human conditions, it seemed inevitable that advances would soon be made using retinal models of disease. The development of CRISPR technology for gene therapy and its increasing potential to selectively target disease-causing nucleotide changes has been rapid. In this chapter, we discuss the currently available CRISPR toolkit and how it has been and can be applied in the future for the treatment of inherited retinal diseases. These blinding conditions have until now had limited opportunity for successful therapeutic intervention, but the discovery of CRISPR has created new hope of achieving such, as we discuss within this chapter.
Collapse
Affiliation(s)
- Ariel Kantor
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom.
| | - Michelle E McClements
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Caroline F Peddle
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Lewis E Fry
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Ahmed Salman
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
23
|
Vedadghavami A, Zhang C, Bajpayee AG. Overcoming negatively charged tissue barriers: Drug delivery using cationic peptides and proteins. NANO TODAY 2020; 34:100898. [PMID: 32802145 PMCID: PMC7425807 DOI: 10.1016/j.nantod.2020.100898] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Negatively charged tissues are ubiquitous in the human body and are associated with a number of common diseases yet remain an outstanding challenge for targeted drug delivery. While the anionic proteoglycans are critical for tissue structure and function, they make tissue matrix dense, conferring a high negative fixed charge density (FCD) that makes drug penetration through the tissue deep zones and drug delivery to resident cells extremely challenging. The high negative FCD of these tissues is now being utilized by taking advantage of electrostatic interactions to create positively charged multi-stage delivery methods that can sequentially penetrate through the full thickness of tissues, create a drug depot and target cells. After decades of work on attempting delivery using strong binding interactions, significant advances have recently been made using weak and reversible electrostatic interactions, a characteristic now considered essential to drug penetration and retention in negatively charged tissues. Here we discuss these advances using examples of negatively charged tissues (cartilage, meniscus, tendons and ligaments, nucleus pulposus, vitreous of eye, mucin, skin), and delve into how each of their structures, tissue matrix compositions and high negative FCDs create barriers to drug entry and explore how charge interactions are being used to overcome these barriers. We review work on tissue targeting cationic peptide and protein-based drug delivery, compare and contrast drug delivery designs, and also present examples of technologies that are entering clinical trials. We also present strategies on further enhancing drug retention within diseased tissues of lower FCD by using synergistic effects of short-range binding interactions like hydrophobic and H-bonds that stabilize long-range charge interactions. As electrostatic interactions are incorporated into design of drug delivery materials and used as a strategy to create properties that are reversible, tunable and dynamic, bio-electroceuticals are becoming an exciting new direction of research and clinical work.
Collapse
Affiliation(s)
- Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
- Department of Mechanical Engineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
24
|
Kurrikoff K, Vunk B, Langel Ü. Status update in the use of cell-penetrating peptides for the delivery of macromolecular therapeutics. Expert Opin Biol Ther 2020; 21:361-370. [PMID: 32938243 DOI: 10.1080/14712598.2021.1823368] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION In this review, recent developments and applications with cell-penetrating peptides (CPP) are discussed. CPPs are widely used tools for the delivery of various macromolecular therapeutics, such as proteins and nucleic acids. AREAS COVERED The current review focuses on recent important advances and reports that demonstrate high clinical and translational potential. Most important clinical developments have occurred with the CPP-drug conjugate approaches that target various protein-protein interactions, and these have been highlighted subsequently. Most of the applications are targeting cancer, but recently, noteworthy advances have taken place in the field of antisense oligonucleotides and muscular dystrophies, lung targeting, and trans-BBB targeting. EXPERT OPINION Successful applications and clinical development with the drug conjugate approaches are discussed. On the other hand, the reasons of why the nanoparticle approaches are not as far in development are analyzed.
Collapse
Affiliation(s)
- Kaido Kurrikoff
- University of Tartu, Institute of Technology, Tartu, Estonia
| | - Birgit Vunk
- University of Tartu, Institute of Technology, Tartu, Estonia
| | - Ülo Langel
- University of Tartu, Institute of Technology, Tartu, Estonia.,Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| |
Collapse
|
25
|
Young CC, Vedadghavami A, Bajpayee AG. Bioelectricity for Drug Delivery: The Promise of Cationic Therapeutics. Bioelectricity 2020; 2:68-81. [PMID: 32803148 DOI: 10.1089/bioe.2020.0012] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biological systems overwhelmingly comprise charged entities generating electrical activity that can have significant impact on biological structure and function. This intrinsic bio-electrical activity can also be harnessed for overcoming the tissue matrix and cell membrane barriers, which have been outstanding challenges for targeted drug delivery, by using rationally designed cationic carriers. The weak and reversible long-range electrostatic interactions with fixed negatively charged groups facilitate electro-diffusive transport of cationic therapeutics through full-tissue thickness to effectively reach intra-tissue, cellular, and intracellular target sites. This article presents a perspective on the promise of using rationally designed cationic biomaterials in targeted drug delivery, the underlying charge-based mechanisms, and bio-transport phenomena while addressing outstanding concerns around toxicity and methods to mitigate them. We also discuss electrically charged drugs that are currently being evaluated in clinical trials and identify areas of further development that have the potential to usher in new treatments.
Collapse
Affiliation(s)
- Cameron C Young
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA.,Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Hyun S, Li L, Yoon KC, Yu J. An amphipathic cell penetrating peptide aids cell penetration of cyclosporin A and increases its therapeutic effect in an in vivo mouse model for dry eye disease. Chem Commun (Camb) 2019; 55:13657-13660. [PMID: 31595891 DOI: 10.1039/c9cc05960a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cell penetrating peptide (CPP), LK-3, causes a ca. 10-fold increase in the cell penetration of cyclosporin A (CsA) at nanomolar concentrations. The results of an in vivo dry eye mouse model demonstrated that a 100-fold lower dose of the CsA/LK-3 complex than that of Restasis® is sufficient to cause the same therapeutic effect.
Collapse
Affiliation(s)
- Soonsil Hyun
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Lan Li
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea.
| | - Kyung Chul Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju 61469, Korea.
| | - Jaehoon Yu
- Department of Chemistry and Education, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
27
|
Gote V, Sikder S, Sicotte J, Pal D. Ocular Drug Delivery: Present Innovations and Future Challenges. J Pharmacol Exp Ther 2019; 370:602-624. [PMID: 31072813 DOI: 10.1124/jpet.119.256933] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/01/2019] [Indexed: 03/08/2025] Open
Abstract
Ocular drug delivery has always been a challenge for ophthalmologists and drug-delivery scientists due to the presence of various anatomic and physiologic barriers. Inimitable static and dynamic ocular barriers not only exclude the entry of xenobiotics but also discourage the active absorption of therapeutic agents. Designing an ideal delivery scheme should include enhanced drug bioavailability and controlled release of drug at the site of action, which can overcome various ocular barriers. Conventional ophthalmic medications include the use of topical eye drops and intravitreal injections of anti-vascular endothelial growth factor agent for treatment of anterior and posterior segment disorders, respectively. Current inventions for anterior ocular segment disorders such as punctum plugs, ocular implants, drug-eluting contact lenses, and ocular iontophoresis represent state-of-the-art inventions for sustained and controlled drug release. Parallel efforts for ocular drug delivery technologies for back of the eye disorders have resulted in the approval of various intravitreal implants. Novel drug-delivery technologies, including nanoparticles, nanomicelles, dendrimers, microneedles, liposomes, and nanowafers, are increasingly studied for anterior and posterior disorders. To achieve patient compliance for back of the eye disorders, novel approaches for noninvasive delivery of potent therapeutic agents are on the rise. In this review article, we discuss past successes, present inventions, and future challenges in ocular drug-delivery technologies. This expert opinion also discusses the future challenges for ocular drug-delivery systems and the clinical translatable potential of nanotechnology from benchtop to bedside.
Collapse
Affiliation(s)
- Vrinda Gote
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| | - Sadia Sikder
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| | - Jeff Sicotte
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| | - Dhananjay Pal
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| |
Collapse
|
28
|
Topical Drug Delivery to the Posterior Segment of the Eye: Addressing the Challenge of Preclinical to Clinical Translation. Pharm Res 2018; 35:245. [PMID: 30374744 PMCID: PMC6208585 DOI: 10.1007/s11095-018-2519-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/03/2018] [Indexed: 12/29/2022]
Abstract
Topical delivery of therapeutics to the posterior segment of the eye remains the “holy grail” of ocular drug delivery. As an example, anti–vascular endothelial growth factor biologics, such as ranibizumab, aflibercept, and bevacizumab, are delivered by intravitreal injection to treat neovascular age-related macular degeneration and, although these drugs have revolutionized treatment of the disease, less invasive alternatives to intravitreal injection are desired. Multiple reports in the literature have demonstrated topical delivery of both small and large molecules to the back of the eye in small animal models. Despite this progress, successful translation to larger species, and ultimately humans, has yet to be demonstrated. Selection of animal models with relevant ocular anatomy and physiology, along with appropriate experimental design, is critical to enable more relevant feasibility assessments and increased probability of successful translation.
Collapse
|
29
|
Pescina S, Ostacolo C, Gomez-Monterrey IM, Sala M, Bertamino A, Sonvico F, Padula C, Santi P, Bianchera A, Nicoli S. Cell penetrating peptides in ocular drug delivery: State of the art. J Control Release 2018; 284:84-102. [PMID: 29913221 DOI: 10.1016/j.jconrel.2018.06.023] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/22/2022]
Abstract
Despite the increasing number of effective therapeutics for eye diseases, their treatment is still challenging due to the presence of effective barriers protecting eye tissues. Cell Penetrating Peptides (CPPs), synthetic and natural short amino acid sequences able to cross cellular membrane thanks to a transduction domain, have been proposed as possible enhancing strategies for ophthalmic delivery. In this review, a general description of CPPs classes, design approaches and proposed cellular uptake mechanisms will be provided to the reader as an introduction to ocular CPPs application, together with an overview of the main problems related to ocular administration. The results obtained with CPPs for the treatment of anterior and posterior segment eye diseases will be then introduced, with a focus on non-invasive or minimally invasive administration, shifting from CPPs capability to obtain intracellular delivery to their ability to cross biological barriers. The problems related to in vitro, ex vivo and in vivo models used to investigate CPPs mediated ocular delivery will be also addressed together with potential ocular toxicity issues.
Collapse
Affiliation(s)
- S Pescina
- Food and Drug Department, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy
| | - C Ostacolo
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - I M Gomez-Monterrey
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - M Sala
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - A Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84084 Fisciano, SA, Italy
| | - F Sonvico
- Food and Drug Department, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy
| | - C Padula
- Food and Drug Department, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy
| | - P Santi
- Food and Drug Department, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy
| | - A Bianchera
- BiopharmanetTEC, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy
| | - S Nicoli
- Food and Drug Department, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy.
| |
Collapse
|
30
|
Talreja D, Cashman SM, Dasari B, Kumar B, Kumar-Singh R. G-quartet oligonucleotide mediated delivery of functional X-linked inhibitor of apoptosis protein into retinal cells following intravitreal injection. Exp Eye Res 2018; 175:20-31. [PMID: 29864441 DOI: 10.1016/j.exer.2018.05.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/31/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022]
Abstract
There is currently no efficient method available for the delivery of full length functional proteins into the cytoplasm of retinal cells in vivo. Historically, the most successful approach for the treatment of retinal diseases has been intravitreal injection of antibodies or recombinant proteins, but this approach is not yet utilized for the delivery of proteins that require intracellular access for a therapeutic effect. Here we describe a platform for the delivery of functional proteins into ganglion cells, photoreceptors and retinal pigment epithelium via intravitreal injection. A nucleolin binding aptamer, AS1411, was biotinylated and complexed with traptavidin and utilized as a platform for the delivery of GFP or X-linked inhibitor of apoptosis (XIAP) proteins by intravitreal injection in BALB/c mice. Retinal sections were analyzed for uptake of proteins in the retina. Apoptosis was induced by intravitreal injection of N-methyl-D-aspartate (NMDA). Retinas were harvested for analysis of TUNEL and caspase 3/7 activity. Intravitreal injection of AS1411-directed GFP or XIAP complexes enabled delivery of these proteins into ganglion cells, photoreceptors and retinal pigment epithelium in vivo. AS1411-XIAP complexes conferred significant protection to cells in the outer and inner nuclear layers following NMDA induced apoptosis. A concomitant decrease in activity of Caspase 3/7 was observed in eyes injected with the AS1411-XIAP complex. In conclusion, AS1411 can be used as a platform for the delivery of therapeutic proteins into retinal cells. This approach can potentially be utilized to introduce a large variety of therapeutically relevant proteins that are previously well characterized to maintain the structural integrity and function of retina, thus, preventing vision loss due to ocular trauma or inherited retinal degeneration.
Collapse
Affiliation(s)
- Deepa Talreja
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Siobhan M Cashman
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Bhanu Dasari
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Binit Kumar
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Rajendra Kumar-Singh
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
31
|
Mandal A, Pal D, Agrahari V, Trinh HM, Joseph M, Mitra AK. Ocular delivery of proteins and peptides: Challenges and novel formulation approaches. Adv Drug Deliv Rev 2018; 126:67-95. [PMID: 29339145 DOI: 10.1016/j.addr.2018.01.008] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 12/21/2017] [Accepted: 01/10/2018] [Indexed: 12/12/2022]
Abstract
The impact of proteins and peptides on the treatment of various conditions including ocular diseases over the past few decades has been advanced by substantial breakthroughs in structural biochemistry, genetic engineering, formulation and delivery approaches. Formulation and delivery of proteins and peptides, such as monoclonal antibodies, aptamers, recombinant proteins and peptides to ocular tissues poses significant challenges owing to their large size, poor permeation and susceptibility to degradation. A wide range of advanced drug delivery systems including polymeric controlled release systems, cell-based delivery and nanowafers are being exploited to overcome the challenges of frequent administration to ocular tissues. The next generation systems integrated with new delivery technologies are anticipated to generate improved efficacy and safety through the expansion of the therapeutic target space. This review will highlight recent advances in formulation and delivery strategies of protein and peptide based biopharmaceuticals. We will also describe the current state of proteins and peptides based ocular therapy and future therapeutic opportunities.
Collapse
|
32
|
Octa-arginine modified lipid emulsions as a potential ocular delivery system for disulfiram: A study of the corneal permeation, transcorneal mechanism and anti-cataract effect. Colloids Surf B Biointerfaces 2017; 160:305-314. [DOI: 10.1016/j.colsurfb.2017.08.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 08/14/2017] [Accepted: 08/21/2017] [Indexed: 12/30/2022]
|
33
|
Cell-penetrating peptide-based non-invasive topical delivery systems. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0373-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Bolhassani A, Jafarzade BS, Mardani G. In vitro and in vivo delivery of therapeutic proteins using cell penetrating peptides. Peptides 2017; 87:50-63. [PMID: 27887988 DOI: 10.1016/j.peptides.2016.11.011] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
The failure of proteins to penetrate mammalian cells or target tumor cells restricts their value as therapeutic tools in a variety of diseases such as cancers. Recently, protein transduction domains (PTDs) or cell penetrating peptides (CPPs) have been shown to promote the delivery of therapeutic proteins or peptides into live cells. The successful delivery of proteins mainly depends on their physicochemical properties. Although, linear cell penetrating peptides are one of the most effective delivery vehicles; but currently, cyclic CPPs has been developed to potently transport bioactive full-length proteins into cells. Up to now, several small protein transduction domains from viral proteins including Tat or VP22 could be fused to other peptides or proteins to entry them in various cell types at a dose-dependent approach. A major disadvantage of PTD-fusion proteins is primary uptake into endosomal vesicles leading to inefficient release of the fusion proteins into the cytosol. Recently, non-covalent complex formation (Chariot) between proteins and CPPs has attracted a special interest to overcome some delivery limitations (e.g., toxicity). Many preclinical and clinical trials of CPP-based delivery are currently under evaluation. Generally, development of more efficient protein transduction domains would significantly increase the potency of protein therapeutics. Moreover, the synergistic or combined effects of CPPs with other delivery systems for protein/peptide drug delivery would promote their therapeutic effects in cancer and other diseases. In this review, we will describe the functions and implications of CPPs for delivering the therapeutic proteins or peptides in preclinical and clinical studies.
Collapse
Affiliation(s)
- Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | | | - Golnaz Mardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
35
|
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: 442] [Impact Index Per Article: 49.1] [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
|
36
|
Pescina S, Sala M, Padula C, Scala MC, Spensiero A, Belletti S, Gatti R, Novellino E, Campiglia P, Santi P, Nicoli S, Ostacolo C. Design and Synthesis of New Cell Penetrating Peptides: Diffusion and Distribution Inside the Cornea. Mol Pharm 2016; 13:3876-3883. [PMID: 27676095 DOI: 10.1021/acs.molpharmaceut.6b00658] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The role of cell penetrating peptides (CPPs) has been challenged in recent years for drug delivery to ocular tissues for the targeting of both anterior and posterior segments. The enhancement of trans-corneal transport for anterior segment targeting is a very important issue possibly leading to important outcomes on efficacy and to the opportunity of topical administration of molecules with unfavorable penetration properties. The aim of the present work was the design and synthesis of new CPPs, deriving from the structure of PEP-1 peptide. Synthesized peptides were labeled with 5-carboxyfluorescein (5-FAM), and their diffusion behavior and distribution inside the cornea were evaluated by a validated ex vivo model and a confocal microscopy approach. Newly synthesized peptides showed similar corneal permeation profiles as PEP-1 (Papp = 0.75 ± 0.56 × 10-6 cm/s), about 2.6-fold higher than 5-FAM (Papp = 0.29 ± 0.08 × 10-6 cm/s) despite the higher molecular weight. Confocal microscopy experiments highlighted the tendency of PEP-1 and its derived peptides to localize in the intercellular space and/or in the plasma membrane. Noteworthy, using penetratin as positive control, a higher trans-corneal permeation (Papp = 6.18 ± 1.46 × 10-6 cm/s) was evidenced together with a diffusion by intracellular route and a different accumulation between wings and basal epithelial cells, probably depending on the stage of cell development. Finally, PEP-1 and pep-7 proved to be safe and well tolerated when tested on human conjuctival cell line.
Collapse
Affiliation(s)
- Silvia Pescina
- Department of Pharmacy, University of Parma , Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Marina Sala
- Department of Pharmacy, University of Salerno , Via G. Paolo II 132, 84084 Fisciano (SA), Italy
| | - Cristina Padula
- Department of Pharmacy, University of Parma , Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Maria Carmina Scala
- Department of Pharmacy, University of Salerno , Via G. Paolo II 132, 84084 Fisciano (SA), Italy
| | - Antonia Spensiero
- Department of Pharmacy, University of Salerno , Via G. Paolo II 132, 84084 Fisciano (SA), Italy
| | - Silvana Belletti
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma , Via Volturno 39, 43126 Parma, Italy
| | - Rita Gatti
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma , Via Volturno 39, 43126 Parma, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II , Via D. Montesano 49, 80131 Napoli, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno , Via G. Paolo II 132, 84084 Fisciano (SA), Italy
| | - Patrizia Santi
- Department of Pharmacy, University of Parma , Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Sara Nicoli
- Department of Pharmacy, University of Parma , Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Naples Federico II , Via D. Montesano 49, 80131 Napoli, Italy
| |
Collapse
|
37
|
Chen C, Liu K, Xu Y, Zhang P, Suo Y, Lu Y, Zhang W, Su L, Gu Q, Wang H, Gu J, Li Z, Xu X. Anti-angiogenesis through noninvasive to minimally invasive intraocular delivery of the peptide CC12 identified by in vivo-directed evolution. Biomaterials 2016; 112:218-233. [PMID: 27768975 DOI: 10.1016/j.biomaterials.2016.09.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 09/20/2016] [Accepted: 09/29/2016] [Indexed: 12/13/2022]
Abstract
Anti-vascular endothelial growth factor (VEGF) therapies are widely used for the treatment of neovascular fundus diseases such as diabetic retinopathy. However, these agents need to be injected intravitreally, because their strong hydrophilicity and high molecular weight prevent them from penetrating cell membranes and complex tissue barriers. Moreover, the repeated injections that are required can cause infection and tissue injury. In this study, we used in vivo-directed evolution phage display technology to identify a novel dodecapeptide, named CC12, with the ability to penetrate the ocular barrier in a noninvasive (via conjunctival sac instillation) or minimally invasive (via retrobulbar injection) manner. KV11, an antiangiogenesis peptide previously demonstrated to inhibit pathological neovascularization in the retina, was then used as a model antiangiogenesis cargo for CC12. We found that conjugation of KV11 peptide with CC12 peptide facilitated the delivery of KV11 to the retina, resulting in significant inhibition of retinal neovascularization development via topical application without tissue toxicity. Collectively, our data of multilevel evaluations demonstrate that CC12 may enable the noninvasive to minimally invasive intraocular delivery of antiangiogenic therapeutics.
Collapse
Affiliation(s)
- Chong Chen
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Kun Liu
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Yupeng Xu
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Pengwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital Affiliated to Medical School of Shanghai Jiao Tong University, Shanghai 200032, PR China
| | - Yan Suo
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Yi Lu
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Wenyuan Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Li Su
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Qing Gu
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Huamao Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital Affiliated to Medical School of Shanghai Jiao Tong University, Shanghai 200032, PR China
| | - Jianren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital Affiliated to Medical School of Shanghai Jiao Tong University, Shanghai 200032, PR China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital Affiliated to Medical School of Shanghai Jiao Tong University, Shanghai 200032, PR China.
| | - Xun Xu
- Department of Ophthalmology, Shanghai Key Laboratory of Fundus Disease, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200080, PR China.
| |
Collapse
|
38
|
In vivo genome editing as a potential treatment strategy for inherited retinal dystrophies. Prog Retin Eye Res 2016; 56:1-18. [PMID: 27623223 DOI: 10.1016/j.preteyeres.2016.09.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/06/2016] [Accepted: 09/08/2016] [Indexed: 12/20/2022]
Abstract
In vivo genome editing represents an emerging field in the treatment of monogenic disorders, as it may constitute a solution to the current hurdles in classic gene addition therapy, which are the low levels and limited duration of transgene expression. Following the introduction of a double strand break (DSB) at the mutational site by highly specific endonucleases, such as TALENs (transcription activator like effector nucleases) or RNA based nucleases (clustered regulatory interspaced short palindromic repeats - CRISPR-Cas), the cell's own DNA repair machinery restores integrity to the DNA strand and corrects the mutant sequence, thus allowing the cell to produce protein levels as needed. The DNA repair happens either through the error prone non-homologous end-joining (NHEJ) pathway or with high fidelity through homology directed repair (HDR) in the presence of a DNA donor template. A third pathway called microhomology mediated endjoining (MMEJ) has been recently discovered. In this review, the authors focus on the different DNA repair mechanisms, the current state of the art tools for genome editing and the particularities of the retina and photoreceptors with regard to in vivo therapeutic approaches. Finally, current attempts in the field of retinal in vivo genome editing are discussed and future directions of research identified.
Collapse
|
39
|
Liu C, Jiang K, Tai L, Liu Y, Wei G, Lu W, Pan W. Facile Noninvasive Retinal Gene Delivery Enabled by Penetratin. ACS APPLIED MATERIALS & INTERFACES 2016; 8:19256-67. [PMID: 27400087 DOI: 10.1021/acsami.6b04551] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Gene delivery to the posterior segment of the eye is severely hindered by the impermeability of defensive barriers; therefore, in clinical settings, genomic medicines are mainly administered by intravitreal injection. We previously found that penetratin could transport the covalently conjugated fluorophore to the fundus oculi by topical instillation. In this study, gene delivery systems enabled by penetratin were designed based on electrostatic binding to target the retina via a noninvasive administration route and prepared with red fluorescent protein plasmid (pRFP) and/or poly(amidoamine) dendrimer of low molecular weight (G3 PAMAM). Formulation optimization, structure confirmation, and characterization were subsequently conducted. Penetratin alone showed limited ability to condense the plasmid but had powerful uptake and transfection by corneal and conjunctival cells. G3 PAMAM was nontoxic to the ocular cells, and when introduced into the penetratin-incorporated complex, the plasmid was condensed more compactly. Therefore, further improved cellular uptake and transfection were observed. After being instilled in the conjunctival sac of rats, the intact complexes penetrated rapidly from the ocular surface into the fundus and resided in the retina for more than 8 h, which resulted in efficient expression of RFP in the posterior segment. Intraocular distribution of the complexes suggested that the plasmids were absorbed into the eyes through a noncorneal pathway during which penetratin played a crucial role. This study provides a facile and friendly approach for intraocular gene delivery and is an important step toward the development of noninvasive gene therapy for posterior segment diseases.
Collapse
Affiliation(s)
- Chang Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Kuan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Lingyu Tai
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University , Shanghai 201203, China
- School of Pharmacy, Shenyang Pharmaceutical University , Shenyang 110016, China
| | - Yu Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Weisan Pan
- School of Pharmacy, Shenyang Pharmaceutical University , Shenyang 110016, China
| |
Collapse
|
40
|
Gautam A, Nanda JS, Samuel JS, Kumari M, Priyanka P, Bedi G, Nath SK, Mittal G, Khatri N, Raghava GPS. Topical Delivery of Protein and Peptide Using Novel Cell Penetrating Peptide IMT-P8. Sci Rep 2016; 6:26278. [PMID: 27189051 PMCID: PMC4870705 DOI: 10.1038/srep26278] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 04/25/2016] [Indexed: 12/21/2022] Open
Abstract
Skin, being the largest organ of the body, is an important site for drug administration. However, most of the drugs have poor permeability and thus drug delivery through the skin is very challenging. In this study, we examined the transdermal delivery capability of IMT-P8, a novel cell-penetrating peptide. We generated IMT-P8-GFP and IMT-P8-KLA fusion constructs and evaluated their internalization into mouse skin after topical application. Our results demonstrate that IMT-P8 is capable of transporting green fluorescent protein (GFP) and proapoptotic peptide, KLA into the skin and also in different cell lines. Interestingly, uptake of IMT-P8-GFP was considerably higher than TAT-GFP in HeLa cells. After internalization, IMT-P8-KLA got localized to the mitochondria and caused significant cell death in HeLa cells signifying an intact biological activity. Further in vivo skin penetration experiments revealed that after topical application, IMT-P8 penetrated the stratum corneum, entered into the viable epidermis and accumulated inside the hair follicles. In addition, both IMT-P8-KLA and IMT-P8-GFP internalized into the hair follicles and dermal tissue of the skin following topical application. These results suggested that IMT-P8 could be a potential candidate to be used as a topical delivery vehicle for various cosmetic and skin disease applications.
Collapse
Affiliation(s)
- Ankur Gautam
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Jagpreet Singh Nanda
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Jesse S Samuel
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Manisha Kumari
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Priyanka Priyanka
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Gursimran Bedi
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Samir K Nath
- Department of Protein Science and Engineering, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Garima Mittal
- Experimental Animal Facility, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Neeraj Khatri
- Experimental Animal Facility, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | | |
Collapse
|
41
|
Leaderer D, Cashman SM, Kumar-Singh R. G-quartet oligonucleotide mediated delivery of proteins into photoreceptors and retinal pigment epithelium via intravitreal injection. Exp Eye Res 2016; 145:380-392. [PMID: 26923800 PMCID: PMC5334003 DOI: 10.1016/j.exer.2016.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 12/27/2022]
Abstract
There is currently no available method to efficiently deliver proteins across the plasma membrane of photoreceptor or retinal pigment epithelium (RPE) cells in vivo. Thus, current clinical application of recombinant proteins in ophthalmology is limited to the use of proteins that perform their biological function extracellularly. The ability to traverse biological membranes would enable the mobilization of a significantly larger number of proteins with previously well characterized properties. Nucleolin is abundantly present on the surface of rapidly dividing cells including cancer cells. Surprisingly, nucleolin is also present on the surface of photoreceptor cell bodies. Here we investigated whether nucleolin can be utilized as a gateway for the delivery of proteins into retinal cells following intravitreal injection. AS1411 is a G-quartet aptamer capable of targeting nucleolin. Subsequent to intravitreal injection, fluorescently labeled AS1411 localized to various retinal cell types including the photoreceptors and RPE. AS1411 linked to streptavidin (a ∼50 kDa protein) via a biotin bridge enabled the uptake of Streptavidin into photoreceptors and RPE. AS1411-Streptavidin conjugate applied topically to the cornea allowed for uptake of the conjugate into the nucleus and cytoplasm of corneal endothelial cells. Clinical relevance of AS1411 as a delivery vehicle was strongly indicated by demonstration of the presence of cell surface nucleolin on the photoreceptors, inner neurons and ganglion cells of human retina. These data support exploration of AS1411 as a means of delivering therapeutic proteins to diseased retina.
Collapse
Affiliation(s)
- Derek Leaderer
- Department of Ophthalmology, Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Siobhan M Cashman
- Department of Ophthalmology, Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Rajendra Kumar-Singh
- Department of Ophthalmology, Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
42
|
Mitra RN, Zheng M, Han Z. Nanoparticle-motivated gene delivery for ophthalmic application. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:160-74. [PMID: 26109528 PMCID: PMC4688250 DOI: 10.1002/wnan.1356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 12/24/2022]
Abstract
Ophthalmic gene therapy is an intellectual and intentional manipulation of desired gene expression into the specific cells of an eye for the treatment of ophthalmic (ocular) genetic dystrophies and pathological conditions. Exogenous nucleic acids such as DNA, small interfering RNA, micro RNA, and so on, are used for the purpose of managing expression of the desired therapeutic proteins in ocular tissues. The delivery of unprotected nucleic acids into the cells is limited because of exogenous and endogenous degradation modalities. Nanotechnology, a promising and sophisticated cutting edge tool, works as a protective shelter for these therapeutic nucleic acids. They can be safely delivered to the required cells in order to modulate anticipated protein expression. To this end, nanotechnology is seen as a potential and promising strategy in the field of ocular gene delivery. This review focused on current nanotechnology modalities and other promising nonviral strategies being used to deliver therapeutic genes in order to treat various devastating ocular diseases.
Collapse
Affiliation(s)
| | - Min Zheng
- Department of Ophthalmology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zongchao Han
- Department of Ophthalmology, University of North Carolina, Chapel Hill, NC 27599, USA
- Carolina Institute for NanoMedicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
43
|
George EM, Mahdi F, Logue OC, Robinson GG, Bidwell GL. Corneal Penetrating Elastin-Like Polypeptide Carriers. J Ocul Pharmacol Ther 2015; 32:163-71. [PMID: 26672799 DOI: 10.1089/jop.2015.0082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Elastin-like polypeptide (ELP) is a bioengineered protein widely applied as a drug carrier due to its biocompatibility and amenability to modification with cell-penetrating peptides (CPPs) and therapeutic agents. The purpose of this study was to determine whether topically applied ELP or CPP-fused ELPs penetrate the corneal barrier. METHODS In vitro binding and cytotoxicity to human corneal epithelial (HCE) cells were determined for ELP or CPP-ELPs. Corneal binding, clearance, and penetration were assessed in a rabbit model following topical application of the fluorescently labeled proteins by quantitative fluorescence imaging and histology. RESULTS ELP bound to HCE cells in vitro, and binding/uptake was enhanced 2- to 3-fold by the addition of CPPs. When applied topically to rabbit eyes, ELP accumulated in the cornea at levels 7.4-fold higher than did an equivalent dose of immunoglobulin G. Both ELP and a CPP-ELP penetrated the corneal epithelium and were detectable in the stroma. Addition of CPPs to ELP, however, did not significantly enhance corneal uptake or penetration in vivo relative to ELP alone. The polypeptides cleared from the cornea over a period of 20-30 min after application, after which cornea levels reached a steady state of 15-30 μg/mL for up to 3 h. CONCLUSIONS The ELP drug carrier can penetrate the corneal epithelium and accumulate in the stroma. Given its amenability for fusion to multiple types of therapeutic agents, ELP has the potential to serve as a drug carrier for topical ocular applications.
Collapse
Affiliation(s)
- Eric M George
- 1 Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,2 Department of Biochemistry, University of Mississippi Medical Center , Jackson, Mississippi
| | - Fakhri Mahdi
- 3 Department of Neurology University of Mississippi Medical Center , Jackson, Mississippi
| | - Omar C Logue
- 3 Department of Neurology University of Mississippi Medical Center , Jackson, Mississippi
| | - Grant G Robinson
- 3 Department of Neurology University of Mississippi Medical Center , Jackson, Mississippi
| | - Gene L Bidwell
- 2 Department of Biochemistry, University of Mississippi Medical Center , Jackson, Mississippi.,3 Department of Neurology University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
44
|
Zulliger R, Conley SM, Naash MI. Non-viral therapeutic approaches to ocular diseases: An overview and future directions. J Control Release 2015; 219:471-487. [PMID: 26439665 PMCID: PMC4699668 DOI: 10.1016/j.jconrel.2015.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/01/2015] [Accepted: 10/02/2015] [Indexed: 12/31/2022]
Abstract
Currently there are no viable treatment options for patients with debilitating inherited retinal degeneration. The vast variability in disease-inducing mutations and resulting phenotypes has hampered the development of therapeutic interventions. Gene therapy is a logical approach, and recent work has focused on ways to optimize vector design and packaging to promote optimized expression and phenotypic rescue after intraocular delivery. In this review, we discuss ongoing ocular clinical trials, which currently use viral gene delivery, but focus primarily on new advancements in optimizing the efficacy of non-viral gene delivery for ocular diseases. Non-viral delivery systems are highly customizable, allowing functionalization to improve cellular and nuclear uptake, bypassing cellular degradative machinery, and improving gene expression in the nucleus. Non-viral vectors often yield transgene expression levels lower than viral counterparts, however their favorable safety/immune profiles and large DNA capacity (critical for the delivery of large ocular disease genes) make their further development a research priority. Recent work on particle coating and vector engineering presents exciting ways to overcome limitations of transient/low gene expression levels, but also highlights the fact that further refinements are needed before use in the clinic.
Collapse
Affiliation(s)
- Rahel Zulliger
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204-5060, United States.
| |
Collapse
|
45
|
Mueller NH, Fogueri U, Pedler MG, Montana K, Petrash JM, Ammar DA. Impact of Subunit Composition on the Uptake of α-Crystallin by Lens and Retina. PLoS One 2015; 10:e0137659. [PMID: 26355842 PMCID: PMC4565700 DOI: 10.1371/journal.pone.0137659] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/19/2015] [Indexed: 11/30/2022] Open
Abstract
Misfolded protein aggregation, including cataract, cause a significant amount of blindness worldwide. α-Crystallin is reported to bind misfolded proteins and prevent their aggregation. We hypothesize that supplementing retina and lens with α-crystallin may help to delay disease onset. The purpose of this study was to determine if αB-crystallin subunits containing a cell penetration peptide (gC-tagged αB-crystallin) facilitate the uptake of wild type αA-crystallin (WT-αA) in lens and retina. Recombinant human αB-crystallin was modified by the addition of a novel cell penetration peptide derived from the gC gene product of herpes simplex virus (gC-αB). Recombinant gC-αB and wild-type αA-crystallin (WT-αA) were purified from E. coli over-expression cultures. After Alexa-labeling of WT-αA, these proteins were mixed at ratios of 1:2, 1:5 and 1:10, respectively, and incubated at 37°C for 4 hours to allow for subunit exchange. Mixed oligomers were subsequently incubated with tissue culture cells or mouse organ cultures. Similarly, crystallin mixtures were injected into the vitreous of rat eyes. At various times after exposure, tissues were harvested and analyzed for protein uptake by confocal microscopy or flow cytometry. Chaperone-like activity assays were performed on α-crystallins ratios showing optimal uptake using chemically-induced or heat induced substrate aggregation assays. As determined by flow cytometry, a ratio of 1:5 for gC-αB to WT-αA was found to be optimal for uptake into retinal pigmented epithelial cells (ARPE-19). Chaperone-like activity assays demonstrated that hetero-oligomeric complex of gC-αB to WT-αA (in 1:5 ratio) retained protein aggregation protection. We observed a significant increase in protein uptake when optimized (gC-αB to WT-αA (1:5 ratio)) hetero-oligomers were used in mouse lens and retinal organ cultures. Increased levels of α-crystallin were found in lens and retina following intravitreal injection of homo- and hetero-oligomers in rats.
Collapse
Affiliation(s)
- Niklaus H. Mueller
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| | - Uma Fogueri
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, Colorado, United States of America
| | - Michelle G. Pedler
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Kameron Montana
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - J. Mark Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, Colorado, United States of America
| | - David A. Ammar
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
46
|
Li H, Tsui TY, Ma W. Intracellular Delivery of Molecular Cargo Using Cell-Penetrating Peptides and the Combination Strategies. Int J Mol Sci 2015; 16:19518-19536. [PMID: 26295227 PMCID: PMC4581311 DOI: 10.3390/ijms160819518] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/11/2015] [Accepted: 07/30/2015] [Indexed: 12/15/2022] Open
Abstract
Cell-penetrating peptides (CPPs) can cross cellular membranes in a non-toxic fashion, improving the intracellular delivery of various molecular cargos such as nanoparticles, small molecules and plasmid DNA. Because CPPs provide a safe, efficient, and non-invasive mode of transport for various cargos into cells, they have been developed as vectors for the delivery of genetic and biologic products in recent years. Most common CPPs are positively charged peptides. While delivering negatively charged molecules (e.g., nucleic acids) to target cells, the internalization efficiency of CPPs is reduced and inhibited because the cationic charges on the CPPs are neutralized through the covering of CPPs by cargos on the structure. Even under these circumstances, the CPPs can still be non-covalently complexed with the negatively charged molecules. To address this issue, combination strategies of CPPs with other typical carriers provide a promising and novel delivery system. This review summarizes the latest research work in using CPPs combined with molecular cargos including liposomes, polymers, cationic peptides, nanoparticles, adeno-associated virus (AAV) and calcium for the delivery of genetic products, especially for small interfering RNA (siRNA). This combination strategy remedies the reduced internalization efficiency caused by neutralization.
Collapse
Affiliation(s)
- Hua Li
- Department of Basic Medical Science, Huzhou University School of Medicine, Huzhou 313000, China.
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Tung Yu Tsui
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Wenxue Ma
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0820, USA.
| |
Collapse
|
47
|
Delivery of nucleic acids and nanomaterials by cell-penetrating peptides: opportunities and challenges. BIOMED RESEARCH INTERNATIONAL 2015; 2015:834079. [PMID: 25883975 PMCID: PMC4391616 DOI: 10.1155/2015/834079] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/18/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022]
Abstract
Many viral and nonviral systems have been developed to aid delivery of biologically active molecules into cells. Among these, cell-penetrating peptides (CPPs) have received increasing attention in the past two decades for biomedical applications. In this review, we focus on opportunities and challenges associated with CPP delivery of nucleic acids and nanomaterials. We first describe the nature of versatile CPPs and their interactions with various types of cargoes. We then discuss in vivo and in vitro delivery of nucleic acids and nanomaterials by CPPs. Studies on the mechanisms of cellular entry and limitations in the methods used are detailed.
Collapse
|
48
|
Koseva NS, Rydz J, Stoyanova EV, Mitova VA. Hybrid protein-synthetic polymer nanoparticles for drug delivery. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 98:93-119. [PMID: 25819277 DOI: 10.1016/bs.apcsb.2014.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Among the most common nanoparticulate systems, the polymeric nanocarriers have a number of key benefits, which give a great choice of delivery platforms. Nevertheless, polymeric nanoparticles possess some limitations that include use of toxic solvents in the production process, polymer degradation, drug leakage outside the diseased tissue, and polymer cytotoxicity. The combination of polymers of biological and synthetic origin is an appealing modern strategy for the production of novel nanocarriers with unprecedented properties. Proteins' interface can play an important role in determining bioactivity and toxicity and gives perspective for future development of the polymer-based nanoparticles. The design of hybrid constructs composed of synthetic polymer and biological molecules such as proteins can be considered as a straightforward tool to integrate a broad spectrum of properties and biofunctions into a single device. This review discusses hybrid protein-synthetic polymer nanoparticles with different structures and levels in complexity and functionality, in view of their applications as drug delivery systems.
Collapse
Affiliation(s)
- Neli S Koseva
- Institute of Polymers, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | - Joanna Rydz
- Institute of Polymers, Bulgarian Academy of Sciences, Sofia, Bulgaria; Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | | | - Violeta A Mitova
- Institute of Polymers, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
49
|
Vasconcelos A, Vega E, Pérez Y, Gómara MJ, García ML, Haro I. Conjugation of cell-penetrating peptides with poly(lactic-co-glycolic acid)-polyethylene glycol nanoparticles improves ocular drug delivery. Int J Nanomedicine 2015; 10:609-31. [PMID: 25670897 PMCID: PMC4315550 DOI: 10.2147/ijn.s71198] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this work, a peptide for ocular delivery (POD) and human immunodeficiency virus transactivator were conjugated with biodegradable poly(lactic-co-glycolic acid) (PGLA)-polyethylene glycol (PEG)-nanoparticles (NPs) in an attempt to improve ocular drug bioavailability. The NPs were prepared by the solvent displacement method following two different pathways. One involved preparation of PLGA NPs followed by PEG and peptide conjugation (PLGA-NPs-PEG-peptide); the other involved self-assembly of PLGA-PEG and the PLGA-PEG-peptide copolymer followed by NP formulation. The conjugation of the PEG and the peptide was confirmed by a colorimetric test and proton nuclear magnetic resonance spectroscopy. Flurbiprofen was used as an example of an anti-inflammatory drug. The physicochemical properties of the resulting NPs (morphology, in vitro release, cell viability, and ocular tolerance) were studied. In vivo anti-inflammatory efficacy was assessed in rabbit eyes after topical instillation of sodium arachidonate. Of the formulations developed, the PLGA-PEG-POD NPs were the smaller particles and exhibited greater entrapment efficiency and more sustained release. The positive charge on the surface of these NPs, due to the conjugation with the positively charged peptide, facilitated penetration into the corneal epithelium, resulting in more effective prevention of ocular inflammation. The in vitro toxicity of the NPs developed was very low; no ocular irritation in vitro (hen's egg test-chorioallantoic membrane assay) or in vivo (Draize test) was detected. Taken together, these data demonstrate that PLGA-PEG-POD NPs are promising vehicles for ocular drug delivery.
Collapse
Affiliation(s)
- Aimee Vasconcelos
- Unit of Synthesis and Biomedical Applications of Peptides, Department of Biomedical Chemistry, Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - Estefania Vega
- Department of Physical Chemistry, Institute of Nanoscience and Nanotechnology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Yolanda Pérez
- Nuclear Magnetic Resonance Unit, IQAC-CSIC, Barcelona, Spain
| | - María J Gómara
- Unit of Synthesis and Biomedical Applications of Peptides, Department of Biomedical Chemistry, Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - María Luisa García
- Department of Physical Chemistry, Institute of Nanoscience and Nanotechnology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Isabel Haro
- Unit of Synthesis and Biomedical Applications of Peptides, Department of Biomedical Chemistry, Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| |
Collapse
|
50
|
Trapani I, Puppo A, Auricchio A. Vector platforms for gene therapy of inherited retinopathies. Prog Retin Eye Res 2014; 43:108-28. [PMID: 25124745 PMCID: PMC4241499 DOI: 10.1016/j.preteyeres.2014.08.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/26/2014] [Accepted: 08/02/2014] [Indexed: 12/20/2022]
Abstract
Inherited retinopathies (IR) are common untreatable blinding conditions. Most of them are inherited as monogenic disorders, due to mutations in genes expressed in retinal photoreceptors (PR) and in retinal pigment epithelium (RPE). The retina's compatibility with gene transfer has made transduction of different retinal cell layers in small and large animal models via viral and non-viral vectors possible. The ongoing identification of novel viruses as well as modifications of existing ones based either on rational design or directed evolution have generated vector variants with improved transduction properties. Dozens of promising proofs of concept have been obtained in IR animal models with both viral and non-viral vectors, and some of them have been relayed to clinical trials. To date, recombinant vectors based on the adeno-associated virus (AAV) represent the most promising tool for retinal gene therapy, given their ability to efficiently deliver therapeutic genes to both PR and RPE and their excellent safety and efficacy profiles in humans. However, AAVs' limited cargo capacity has prevented application of the viral vector to treatments requiring transfer of genes with a coding sequence larger than 5 kb. Vectors with larger capacity, i.e. nanoparticles, adenoviral and lentiviral vectors are being exploited for gene transfer to the retina in animal models and, more recently, in humans. This review focuses on the available platforms for retinal gene therapy to fight inherited blindness, highlights their main strengths and examines the efforts to overcome some of their limitations.
Collapse
Affiliation(s)
- Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Agostina Puppo
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy; Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy.
| |
Collapse
|