1
|
Dias MF, Cruz-Cazarim ELC, Pittella F, Baião A, Pacheco AC, Sarmento B, Fialho SL. Co-delivery of antioxidants and siRNA-VEGF: promising treatment for age-related macular degeneration. Drug Deliv Transl Res 2025:10.1007/s13346-024-01772-x. [PMID: 39751765 DOI: 10.1007/s13346-024-01772-x] [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] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
Current treatments for retinal disorders are anti-angiogenic agents, laser photocoagulation, and photodynamic therapies. These conventional treatments focus on reducing abnormal blood vessel formation in the retina, which, in a low-oxygen environment, can lead to harmful proliferation of endothelial cells. This results in dysfunctional, leaky blood vessels that cause retinal edema, hemorrhage, and vision loss. Age-related Macular Degeneration is a primary cause of vision loss and blindness in the elderly, impacting around 20% of those over 50 years old. This complex disease is also closely related to oxidative stress in retina. In this review, we explore the challenge of treating retinal diseases, alternatives and possibilities of enhancing the effectiveness of therapies using co-delivery systems containing both antiangiogenic and antioxidant therapeutic agents. Despite recent proposals potential, the lack of extensive clinical studies on the long-term outcomes and optimal combinations of therapies means that the full risk profile and effectiveness of combined therapy are not yet completely understood. These factors must be carefully considered and managed by healthcare providers to optimize treatment outcomes and ensure patient safety.
Collapse
Affiliation(s)
- Marina F Dias
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, CEP 30510-010, Minas Gerais, Brazil
| | - Estael L C Cruz-Cazarim
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Juiz de Fora, Juiz de Fora, CEP 36036-900, Minas Gerais, Brazil
| | - Frederico Pittella
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Juiz de Fora, Juiz de Fora, CEP 36036-900, Minas Gerais, Brazil
| | - Ana Baião
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, ICBAS, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ana Catarina Pacheco
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- CESPU-IUCS, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Bruno Sarmento
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- CESPU-IUCS, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Silvia L Fialho
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, CEP 30510-010, Minas Gerais, Brazil.
| |
Collapse
|
2
|
Ebrahimi N, Manavi MS, Nazari A, Momayezi A, Faghihkhorasani F, Rasool Riyadh Abdulwahid AH, Rezaei-Tazangi F, Kavei M, Rezaei R, Mobarak H, Aref AR, Fang W. Nano-scale delivery systems for siRNA delivery in cancer therapy: New era of gene therapy empowered by nanotechnology. ENVIRONMENTAL RESEARCH 2023; 239:117263. [PMID: 37797672 DOI: 10.1016/j.envres.2023.117263] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
RNA interference (RNAi) is a unique treatment approach used to decrease a disease's excessive gene expression, including cancer. SiRNAs may find and destroy homologous mRNA sequences within the cell thanks to RNAi processes. However, difficulties such poor cellular uptake, off-target effects, and susceptibility to destruction by serum nucleases in the bloodstream restrict the therapeutic potential of siRNAs. Since some years ago, siRNA-based therapies have been in the process of being translated into the clinic. Therefore, the primary emphasis of this work is on sophisticated nanocarriers that aid in the transport of siRNA payloads, their administration in combination with anticancer medications, and their use in the treatment of cancer. The research looks into molecular manifestations, difficulties with siRNA transport, the design and development of siRNA-based delivery methods, and the benefits and drawbacks of various nanocarriers. The trapping of siRNA in endosomes is a challenge for the majority of delivery methods, which affects the therapeutic effectiveness. Numerous techniques for siRNA release, including as pH-responsive release, membrane fusion, the proton sponge effect, and photochemical disruption, have been studied to overcome this problem. The present state of siRNA treatments in clinical trials is also looked at in order to give a thorough and systematic evaluation of siRNA-based medicines for efficient cancer therapy.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Iran
| | | | - Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Amirali Momayezi
- School of Chemical Engineering, Iran University of Science, and Technology, Tehran, Iran
| | | | | | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Mohammed Kavei
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Roya Rezaei
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Halimeh Mobarak
- Clinical Pathologist, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - Wei Fang
- Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Kang H, Ga YJ, Kim SH, Cho YH, Kim JW, Kim C, Yeh JY. Small interfering RNA (siRNA)-based therapeutic applications against viruses: principles, potential, and challenges. J Biomed Sci 2023; 30:88. [PMID: 37845731 PMCID: PMC10577957 DOI: 10.1186/s12929-023-00981-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023] Open
Abstract
RNA has emerged as a revolutionary and important tool in the battle against emerging infectious diseases, with roles extending beyond its applications in vaccines, in which it is used in the response to the COVID-19 pandemic. Since their development in the 1990s, RNA interference (RNAi) therapeutics have demonstrated potential in reducing the expression of disease-associated genes. Nucleic acid-based therapeutics, including RNAi therapies, that degrade viral genomes and rapidly adapt to viral mutations, have emerged as alternative treatments. RNAi is a robust technique frequently employed to selectively suppress gene expression in a sequence-specific manner. The swift adaptability of nucleic acid-based therapeutics such as RNAi therapies endows them with a significant advantage over other antiviral medications. For example, small interfering RNAs (siRNAs) are produced on the basis of sequence complementarity to target and degrade viral RNA, a novel approach to combat viral infections. The precision of siRNAs in targeting and degrading viral RNA has led to the development of siRNA-based treatments for diverse diseases. However, despite the promising therapeutic benefits of siRNAs, several problems, including impaired long-term protein expression, siRNA instability, off-target effects, immunological responses, and drug resistance, have been considerable obstacles to the use of siRNA-based antiviral therapies. This review provides an encompassing summary of the siRNA-based therapeutic approaches against viruses while also addressing the obstacles that need to be overcome for their effective application. Furthermore, we present potential solutions to mitigate major challenges.
Collapse
Affiliation(s)
- Hara Kang
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea
| | - Yun Ji Ga
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea
| | - Soo Hyun Kim
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea
| | - Young Hoon Cho
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea
| | - Jung Won Kim
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea
- Convergence Research Center for Insect Vectors, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea
| | - Chaeyeon Kim
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea
| | - Jung-Yong Yeh
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea.
- Research Institute for New Drug Development, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea.
- Convergence Research Center for Insect Vectors, Incheon National University, Academy-Ro 119, Yeonsu-Gu, Incheon, 22012, South Korea.
- KU Center for Animal Blood Medical Science, College of Veterinary Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, South Korea.
| |
Collapse
|
4
|
Corydon IJ, Fabian-Jessing BK, Jakobsen TS, Jørgensen AC, Jensen EG, Askou AL, Aagaard L, Corydon TJ. 25 years of maturation: A systematic review of RNAi in the clinic. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:469-482. [PMID: 37583575 PMCID: PMC10424002 DOI: 10.1016/j.omtn.2023.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The year 2023 marks the 25th anniversary of the discovery of RNAi. RNAi-based therapeutics enable sequence-specific gene knockdown by eliminating target RNA molecules through complementary base-pairing. A systematic review of published and ongoing clinical trials was performed. Web of Science, PubMed, and Embase were searched from January 1, 1998, to December 30, 2022 for clinical trials using RNAi. Following inclusion, data from the articles were extracted according to a predefined protocol. A total of 90 trials published in 81 articles were included. In addition, ongoing clinical trials were retrieved from ClinicalTrials.gov, resulting in the inclusion of 48 trials. We investigated how maturation of RNAi-based therapeutics and developments in delivery platforms, administration routes, and potential targets shape the current landscape of clinically applied RNAi. Notably, most contemporary clinical trials used either N-acetylgalactosamine delivery and subcutaneous administration or lipid nanoparticle delivery and intravenous administration. In conclusion, RNAi therapeutics have gained great momentum during the past decade, resulting in five approved therapeutics targeting the liver for treatment of severe diseases, and the trajectory depicted by the ongoing trials emphasizes that even more RNAi-based medicines also targeting extra-hepatic tissues are likely to be available in the years to come.
Collapse
Affiliation(s)
- Ida Juhl Corydon
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Bjørn Kristensen Fabian-Jessing
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, Aarhus N, Denmark
| | - Thomas Stax Jakobsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, Aarhus N, Denmark
| | | | - Emilie Grarup Jensen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Anne Louise Askou
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, Aarhus N, Denmark
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Thomas Juhl Corydon
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, Aarhus N, Denmark
| |
Collapse
|
5
|
Supe S, Upadhya A, Tripathi S, Dighe V, Singh K. Liposome-polyethylenimine complexes for the effective delivery of HuR siRNA in the treatment of diabetic retinopathy. Drug Deliv Transl Res 2023; 13:1675-1698. [PMID: 36630075 DOI: 10.1007/s13346-022-01281-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 01/12/2023]
Abstract
Diabetic retinopathy (DR) is a vision-impairing complication of diabetes, damaging the retinal microcirculatory system. Overexpression of VEGF (vascular endothelial growth factor) is implicated in the pathogenesis of DR. Human antigen R (HuR) is an RNA-binding protein that favorably regulates VEGF protein expression by binding to VEGF-encoding mRNA. Downregulating HuR via RNA interference strategies using small interfering RNAs (siRNAs) may constitute a novel therapeutic method for preventing VEGF protein overexpression in DR. Delivery of siRNAs to the cellular cytoplasm can be facilitated by cationic peptides or polymers and lipids. In this study, a cationic polymer (polyethylenimine (PEI)) and lipid nanoparticles (liposomes) were co-formulated with siRNA to form lipopolyplexes (LPPs) for the delivery of HuR siRNA. LPPs-siRNA were analyzed for size, zeta potential, serum stability, RNase stability, heparin stability, toxicity, and siRNA encapsulation efficiency. Cellular uptake, downregulation of the target HuR (mRNA and protein), and associated VEGF protein were used to demonstrate the biological efficacy of the LPPs-HuR siRNA, in vitro (human ARPE-19 cells), and in vivo (Wistar rats). In vivo efficacy study was performed by injecting LPPs-HuR siRNA formulations into the eye of streptozotocin (STZ)-induced diabetic rats after the development of retinopathy. Our findings demonstrated that high retinal HuR and VEGF levels observed in the eyes of untreated STZ rats were lowered after LPPs-HuR siRNA administration. Our observations indicate that intravitreal treatment with HuR siRNA is a promising option for DR using LPPs as delivery agents.
Collapse
Affiliation(s)
- Shibani Supe
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, 400056, India
| | - Archana Upadhya
- Humera Khan College of Pharmacy, HK College Campus, Oshiwara, Jogeshwari (West), Mumbai, Maharashtra, 400102, India
| | - Santosh Tripathi
- Bombay Veterinary College, Sindhu Nagar, Parel Village, Parel, Mumbai, Maharashtra, 400012, India
| | - Vikas Dighe
- National Centre for Preclinical Reproductive and Genetic Toxicology, ICMR-National Institute for Research in Reproductive and Child Health, J.M.Street, Parel, Mumbai, Maharashtra, 400012, India.
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
6
|
Agarwal A, Sarma DK, Chaurasia D, Maan HS. Novel molecular approaches to combat vectors and vector-borne viruses: Special focus on RNA interference (RNAi) mechanisms. Acta Trop 2022; 233:106539. [PMID: 35623398 DOI: 10.1016/j.actatropica.2022.106539] [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: 04/19/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022]
Abstract
Vector-borne diseases, such as dengue, chikungunya, zika, yellow fever etc pose significant burden among the infectious diseases globally, especially in tropical and sub-tropical regions. Globalization, deforestation, urbanization, climate change, uncontrolled population growth, inadequate waste management and poor vector-management infrastructure have all contributed to the expansion of vector habitats and subsequent increase in vector-borne diseases throughout the world. Conventional vector control methods, such as use of insecticides, have significant negative environmental repercussions in addition to developing resistance in vectors. Till date, a very few vaccines or antiviral therapies have been approved for the treatment of vector borne diseases. In this review, we have discussed emerging molecular approaches like CRISPR (clustered regularly interspaced short palindromic repeats)/Cas-9, sterile insect technique (SIT), release of insects carrying a dominant lethal (RIDL), Wolbachia (virus transmission blocking) and RNA interference (RNAi) to combat vector and vector-borne viruses. Due to the extensive advancements in RNAi research, a special focus has been given on its types, biogenesis, mechanism of action, delivery and experimental studies evaluating their application as anti-mosquito and anti-viral agent. These technologies appear to be highly promising in terms of contributing to vector control and antiviral drug development, and hence can be used to reduce global vector and vector-borne disease burden.
Collapse
Affiliation(s)
- Ankita Agarwal
- State Virology Laboratory, Department of Microbiology, Gandhi Medical College, Bhopal 462001, Madhya Pradesh, India.
| | - Devojit Kumar Sarma
- ICMR-National Institute for Research in Environmental Health, Bhopal 462030, Madhya Pradesh, India
| | - Deepti Chaurasia
- State Virology Laboratory, Department of Microbiology, Gandhi Medical College, Bhopal 462001, Madhya Pradesh, India
| | - Harjeet Singh Maan
- State Virology Laboratory, Department of Microbiology, Gandhi Medical College, Bhopal 462001, Madhya Pradesh, India
| |
Collapse
|
7
|
Moreno-Montañés J, Bleau AM, Martínez T, Vargas B, González MV, Jiménez AI. siRNA Therapeutics in Ocular Diseases. Methods Mol Biol 2021; 2282:417-442. [PMID: 33928588 DOI: 10.1007/978-1-0716-1298-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The field of RNAi therapeutics has quickly adapted to the treatment of ocular diseases. Although the eye provides a unique system for the delivery of siRNAs, its complex structure and composition fostered the development of novel strategies for efficient gene silencing in the target compartment. Moreover, anterior and posterior segments differ in their multiple drug barriers and clearance mechanisms. This chapter summarizes the recent achievements in terms of routes of administration, chemical modifications, and delivery systems for siRNAs that specifically apply to eye disorders. Methods employed for siRNA detection/quantitation in ocular tissues are also described, together with safety concerns that need to be addressed to fulfill regulatory requirements of new drug approval. Even though RNAi therapies for ocular diseases have not yet translated into patient care, we document herein the rising number of candidate drugs currently under preclinical or clinical development.
Collapse
|
8
|
Delivery of siRNA to the Eye: Protocol for a Feasibility Study to Assess Novel Delivery System for Topical Delivery of siRNA Therapeutics to the Ocular Surface. Methods Mol Biol 2021. [PMID: 33928589 DOI: 10.1007/978-1-0716-1298-9_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Drug delivery to the eye remains a real challenge due to the presence of ocular anatomical barriers and physiological protective mechanisms. The lack of effective siRNA delivery mechanism has hampered the real potential of RNAi therapy, but recent literature suggests that nanocarrier systems show great promise in enhancing siRNA bioavailability and reducing the need for repeated intraocular injections. A diverse range of materials are under exploration worldwide, including natural and synthetic polymers, liposomes, peptides, and dendrimeric nanomaterials. This chapter describes a simple workflow for feasibility assessment of a proposed ocular surface siRNA delivery system. Gel retardation assay is used for investigation of optimal siRNA to carrier loading ratio. Fluorescent siRNA allows for initial in vitro testing of cellular uptake to corneal epithelial cells and investigation of in vivo siRNA delivery into mouse cornea by live animal imaging and fluorescence microscopy.
Collapse
|
9
|
Neuroprotective Effect of siRNA Entrapped in Hyaluronic Acid-Coated Lipoplexes by Intravitreal Administration. Pharmaceutics 2021; 13:pharmaceutics13060845. [PMID: 34200993 PMCID: PMC8226864 DOI: 10.3390/pharmaceutics13060845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 12/19/2022] Open
Abstract
Since the possibility of silencing specific genes linked to retinal degeneration has become a reality with the use of small interfering RNAs (siRNAs), this technology has been widely studied to promote the treatment of several ocular diseases. Despite recent advances, the clinical success of gene silencing in the retina is significantly reduced by inherent anatomical and physiological ocular barriers, and new strategies are required to achieve intraocular therapeutic effectiveness. In this study, we developed lipoplexes, prepared with sodium alginate as an adjuvant and strategically coated with hyaluronic acid (HA-LIP), and investigated the potential neuroprotective effect of these systems in a retinal light damage model. Successful functionalization of the lipoplexes with hyaluronic acid was indicated in the dynamic light scattering and transmission electron microscopy results. Moreover, these HA-LIP nanoparticles were able to protect and deliver siRNA molecules targeting caspase-3 into the retina. After retinal degeneration induced by high light exposure, in vitro and in vivo quantitative reverse transcription-PCR (RT-qPCR) assays demonstrated significant inhibition of caspase-3 expression by HA-LIP. Furthermore, these systems were shown to be safe, as no evidence of retinal toxicity was observed by electroretinography, clinical evaluation or histology.
Collapse
|
10
|
Supe S, Upadhya A, Singh K. Role of small interfering RNA (siRNA) in targeting ocular neovascularization: A review. Exp Eye Res 2020; 202:108329. [PMID: 33198953 DOI: 10.1016/j.exer.2020.108329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/30/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022]
Abstract
Ocular neovascularization (NV) plays a central role in the pathogenesis of various ocular diseases including diabetic retinopathy, age-related macular degeneration, retinoblastoma, retinitis pigmentosa and may lead to loss of vision if not controlled in time. Several clinical trials elucidate the central role of vascular endothelial growth factor (VEGF) in the pathogenesis of the ocular neovascularization. The advent and extensive use of ocular anti-VEGF therapy heralded a new age in the treatment of retinal vascular and exudative diseases. RNA interference (RNAi) can be used to inhibit the in-vitro and in-vivo expression of specific genes and thus provides an extremely useful method for investigating gene activity with minimal toxicity. siRNA targeting VEGF overcomes many drawbacks associated with the conventional treatment available for the treatment of ocular neovascularization. However, delivery methods that protect the siRNA against degradation and are appropriate for long-term care will help increase the effectiveness of RNAi-based anti-VEGF ocular therapies. Several nanotechnology approaches have been explored by formulation scientists for delivery of siRNA to the eye; targeting particularly VEGF for the treatment of NV. This review mainly focuses on current updates in various pre-clinical and clinical siRNA strategies for targeting VEGF involved in the development of ocular neovascularization.
Collapse
Affiliation(s)
- Shibani Supe
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, 400056, Maharashtra, India
| | - Archana Upadhya
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, 400056, Maharashtra, India
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, 400056, Maharashtra, India.
| |
Collapse
|
11
|
Begum G, Leigh T, Courtie E, Moakes R, Butt G, Ahmed Z, Rauz S, Logan A, Blanch RJ. Rapid assessment of ocular drug delivery in a novel ex vivo corneal model. Sci Rep 2020; 10:11754. [PMID: 32678110 PMCID: PMC7366725 DOI: 10.1038/s41598-020-68254-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/05/2020] [Indexed: 12/29/2022] Open
Abstract
Drug delivery by topical application has higher patient acceptance and lower morbidity than intraocular injection, but many ophthalmic treatments are unable to enter the eye or reach the posterior segment after topical application. The first stage towards posterior segment delivery after topical application is ocular surface penetration and existing models are in vivo or use large quantities of tissue. We therefore developed a novel ex vivo model using discs of porcine and human cornea and sclera (5 mm diameter) to assess penetration of a candidate neuroprotective siRNA. siRNA against caspase 2 or control solutions of known penetrance were applied to the corneal epithelial surface and trans-corneal penetration and corneal adsorbance measured at fixed time points. To demonstrate that leakage did not occur, we applied dextran blue, which should not penetrate the intact cornea and did not do so in our model. Fluorescein penetration (0.09%) was less than rhodamine B (6.98%) at 60 min. siCASP2 penetration was 0.01% by 60 min. When the applied siCASP2 was washed off after 2 min, (representing lacrimal drainage) 0.071% penetrated porcine cornea by 60 min and 0.0002% penetrated human cornea and 0.001% penetrated human sclera. Our ex vivo model rapidly and cost-effectively assesses transcorneal penetration of candidate topical therapies, allowing rates of trans-corneal penetration for potential therapies such as siRNA to be evaluated with small quantities of human or animal tissue.
Collapse
Affiliation(s)
- Ghazala Begum
- miRNA Diagnostics, Birmingham, UK.,Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
| | - Thomas Leigh
- School of Chemistry, University of Birmingham, Birmingham, UK
| | - Ella Courtie
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
| | - Richard Moakes
- School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Gibran Butt
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, University of Birmingham, Birmingham, UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Saaeha Rauz
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, University of Birmingham, Birmingham, UK
| | - Ann Logan
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
| | - Richard J Blanch
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK. .,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK. .,Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, UK. .,Department of Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| |
Collapse
|
12
|
Humphreys SC, Thayer MB, Campbell J, Chen WLK, Adams D, Lade JM, Rock BM. Emerging siRNA Design Principles and Consequences for Biotransformation and Disposition in Drug Development. J Med Chem 2020; 63:6407-6422. [PMID: 32352779 DOI: 10.1021/acs.jmedchem.9b01839] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
After two decades teetering at the intersection of laboratory tool and therapeutic reality, with two siRNA drugs now clinically approved, this modality has finally come into fruition. Consistent with other emerging modalities, initial proof-of-concept efforts concentrated on coupling pharmacologic efficacy with desirable safety profiles. Consequently, thorough investigations of siRNA absorption, distribution, metabolism, and excretion (ADME) properties are lacking. Advancing ADME knowledge will aid establishment of in vitro-in vivo correlations and pharmacokinetic-pharmacodynamic relationships to optimize candidate selection through discovery and translation. Here, we outline the emerging siRNA design principles and discuss the consequences for siRNA disposition and biotransformation. We propose a conceptual framework for siRNA ADME evaluation, contextualizing the site of biotransformation product formation with PK-PD modulation, and end with a discussion around safety and regulatory considerations and future directions for this modality.
Collapse
Affiliation(s)
- Sara C Humphreys
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Mai B Thayer
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Jabbar Campbell
- Neuroscience Department, Amgen Research, 360 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Wen Li Kelly Chen
- Comparative Biology and Safety Sciences Department, Amgen Research, 360 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Dan Adams
- Comparative Biology and Safety Sciences Department, Amgen Research, 360 Binney Street, Cambridge, Massachusetts 02141, United States
| | - Julie M Lade
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| | - Brooke M Rock
- Pharmacokinetics and Drug Metabolism Department, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
13
|
Jiang K, Hu Y, Gao X, Zhan C, Zhang Y, Yao S, Xie C, Wei G, Lu W. Octopus-like Flexible Vector for Noninvasive Intraocular Delivery of Short Interfering Nucleic Acids. NANO LETTERS 2019; 19:6410-6417. [PMID: 31442373 DOI: 10.1021/acs.nanolett.9b02596] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Gene therapy is promising for chronic posterior ocular diseases, which are causal factors for severe vision impairment and even blindness worldwide. However, the inherent absorption barriers of the eye restrict intraocular delivery of therapeutic nucleic acids via topical instillation. Safe and efficient nonviral vectors for ocular gene therapy are still unmet clinical desires. Herein, an octopus-like flexible multivalent penetratin (MVP) was designed to facilitate condensation and delivery of therapeutic nucleic acids using multiarm polyethylene glycol (PEG) as a core and conjugating penetratin at each end of the PEG arms as outspread tentacles. Among the MVPs, 8-valent penetratin (8VP) stably compacted nucleic acids into positively charged polyplexes smaller than 100 nm, promoting cellular uptake efficiency (approaching 100%) and transfection rate (over 75%). After being instilled into the conjunctival sac, 8VP enabled rapid (<10 min) and prolonged (>6 h) distribution of nucleic acids in the retina via a noncorneal pathway. In a retinoblastoma-bearing mice model, topical instillation of 8VP/siRNA efficiently inhibited the protein expression of intraocular tumor without toxicity. MVP is advantageous over the commercial transfection reagent in safety and efficiency, and therefore provides a promising vector for noninvasive intraocular gene delivery.
Collapse
Affiliation(s)
- Kuan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Yang Hu
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Xin Gao
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Changyou Zhan
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
- Department of Pharmacology, School of Basic Medical Sciences , Fudan University , Shanghai 200032 , China
| | - Yanyu Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Shengyu Yao
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , China
| | - Cao Xie
- 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
| |
Collapse
|
14
|
Current Transport Systems and Clinical Applications for Small Interfering RNA (siRNA) Drugs. Mol Diagn Ther 2019; 22:551-569. [PMID: 29926308 DOI: 10.1007/s40291-018-0338-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small interfering RNAs (siRNAs) are an attractive new agent with potential as a therapeutic tool because of its ability to inhibit specific genes for many conditions, including viral infections and cancers. However, despite this potential, many challenges remain, including off-target effects, difficulties with delivery, immune responses, and toxicity. Traditional genetic vectors do not guarantee that siRNAs will silence genes in vivo. Rational design strategies, such as chemical modification, viral vectors, and non-viral vectors, including cationic liposomes, polymers, nanocarriers, and bioconjugated siRNAs, provide important opportunities to overcome these challenges. We summarize the results of research into vector delivery of siRNAs as a therapeutic agent from their design to clinical trials in ophthalmic diseases, cancers, respiratory diseases, and liver virus infections. Finally, we discuss the current state of siRNA delivery methods and the need for greater understanding of the requirements.
Collapse
|
15
|
Aghamiri S, Jafarpour A, Gomari MM, Ghorbani J, Rajabibazl M, Payandeh Z. siRNA nanotherapeutics: a promising strategy for anti‐HBV therapy. IET Nanobiotechnol 2019; 13:457-463. [PMCID: PMC8676379 DOI: 10.1049/iet-nbt.2018.5286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/18/2018] [Accepted: 01/28/2019] [Indexed: 07/31/2023] Open
Abstract
Chronic hepatitis B (CHB) is the most common cause of hepatocellular carcinoma (HCC) and liver cirrhosis worldwide. In spite of the numerous advances in the treatment of CHB, drugs and vaccines have failed because of many factors like complexity, resistance, toxicity, and heavy cost. New RNA interference (RNAi)‐based technologies have developed innovative strategies to target Achilles' heel of the several hazardous diseases involving cancer, some genetic disease, autoimmune illnesses, and viral disorders particularly hepatitis B virus (HBV) infections. Naked siRNA delivery has serious challenges including failure to cross the cell membrane, susceptibility to the enzymatic digestion, and excretion by renal filtration, which ideally can be addressed by nanoparticle‐mediated delivery systems. cccDNA formation is a significant problem in obtaining HBV infections complete cure because of strength, durability, and lack of proper immune response. Nano‐siRNA drugs have a great potential to address this problem by silencing specific genes which are involved in cccDNA formation. In this article, the authors describe siRNA nanocarrier‐mediated delivery systems as a promising new strategy for HBV infections therapy. Simultaneously, the authors completely represent the clinical trials which use these strategies for treatment of the HBV infections.
Collapse
Affiliation(s)
- Shahin Aghamiri
- Student research committeeDepartment of Medical BiotechnologySchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Ali Jafarpour
- Students' Scientific Research CenterVirology DivisionDepartment of PathobiologySchool of Public HealthTehran University of Medical SciencesTehranIran
| | | | - Jaber Ghorbani
- Department of Medical BiotechnologySchool of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Masoumeh Rajabibazl
- Department of Clinical BiochemistryFaculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zahra Payandeh
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
16
|
Corneal chemical burn treatment through a delivery system consisting of TGF-β 1 siRNA: in vitro and in vivo. Drug Deliv Transl Res 2018; 8:1127-1138. [PMID: 29869292 DOI: 10.1007/s13346-018-0546-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chemical burns are major causes of corneal blindness. Transforming growth factor beta-1 (TGFβ1) plays an important role in induction of corneal inflammation-related-fibrosis leading to the blindness. Here, a topical delivery system consisting anti-fibrotic TGF-β1 siRNA, an inflammatory suppressing gene, was designed for treatment of corneal injuries. TGF-β1 siRNA loaded in nanoparticles (NPs) made up of polyethyleneimine polymer demonstrated high fibroblast transfection efficiency. Moreover, TGF-β1 and PDGF genes and ECM deposition were suppressed in isolated human corneal fibroblasts. NPs inhibited proliferation and transformation of fibroblasts to myofibroblasts by S-phase arrest and α-SMA suppression in vitro, respectively. The mentioned finding was also confirmed in vivo, addressing high wound-healing potential of prepared gene delivery system which was superior to conventional betamethasone treatment. Besides, CD4+ and α-SMA antibody staining showed inhibited angiogenesis and myofibroblast accumulation in treated corneas. This study opens a new way for treating corneal fibrosis through topical siRNA delivery.
Collapse
|
17
|
Dillinger AE, Guter M, Froemel F, Weber GR, Perkumas K, Stamer WD, Ohlmann A, Fuchshofer R, Breunig M. Intracameral Delivery of Layer-by-Layer Coated siRNA Nanoparticles for Glaucoma Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1803239. [PMID: 30353713 PMCID: PMC6599181 DOI: 10.1002/smll.201803239] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/02/2018] [Indexed: 05/12/2023]
Abstract
Glaucoma is the second leading cause of blindness worldwide, often associated with elevated intraocular pressure. Connective tissue growth factor (CTGF) is a mediator of pathological effects in the trabecular meshwork (TM) and Schlemm's canal (SC). A novel, causative therapeutic concept which involves the intracameral delivery of small interfering RNA against CTGF is proposed. Layer-by-layer coated nanoparticles of 200-260 nm with a final layer of hyaluronan (HA) are developed. The HA-coating should provide the nanoparticles sufficient mobility in the extracellular matrix and allow for binding to TM and SC cells via CD44. By screening primary TM and SC cells in vitro, in vivo, and ex vivo, the validity of the concept is confirmed. CD44 expression is elevated in glaucomatous versus healthy cells by about two- to sixfold. CD44 is significantly involved in the cellular uptake of HA-coated nanoparticles. Ex vivo organ culture of porcine, murine, and human eyes demonstrates up to threefold higher accumulation of HA compared to control nanoparticles and much better penetration into the target tissue. Gene silencing in primary human TM cells results in a significant reduction of CTGF expression. Thus, HA-coated nanoparticles combined with RNA interference may provide a potential strategy for glaucoma therapy.
Collapse
Affiliation(s)
- Andrea E Dillinger
- Department of Human Anatomy and Embryology, University Regensburg, Universitaetsstrasse 31, 93040, Regensburg, Germany
| | - Michaela Guter
- Department of Pharmaceutical Technology, University Regensburg, Universitaetsstrasse 31, 93040, Regensburg, Germany
| | - Franziska Froemel
- Department of Human Anatomy and Embryology, University Regensburg, Universitaetsstrasse 31, 93040, Regensburg, Germany
| | - Gregor R Weber
- Department of Human Anatomy and Embryology, University Regensburg, Universitaetsstrasse 31, 93040, Regensburg, Germany
| | - Kristin Perkumas
- Department of Ophthalmology, Duke University, 2351 Erwin Road, Durham, NC, 27710, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, 2351 Erwin Road, Durham, NC, 27710, USA
| | - Andreas Ohlmann
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, 80336, Munich, Germany
| | - Rudolf Fuchshofer
- Department of Human Anatomy and Embryology, University Regensburg, Universitaetsstrasse 31, 93040, Regensburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University Regensburg, Universitaetsstrasse 31, 93040, Regensburg, Germany
| |
Collapse
|
18
|
Qureshi A, Tantray VG, Kirmani AR, Ahangar AG. A review on current status of antiviral siRNA. Rev Med Virol 2018; 28:e1976. [PMID: 29656441 PMCID: PMC7169094 DOI: 10.1002/rmv.1976] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/18/2018] [Accepted: 02/12/2018] [Indexed: 01/12/2023]
Abstract
Viral diseases like influenza, AIDS, hepatitis, and Ebola cause severe epidemics worldwide. Along with their resistant strains, new pathogenic viruses continue to be discovered so creating an ongoing need for new antiviral treatments. RNA interference is a cellular gene‐silencing phenomenon in which sequence‐specific degradation of target mRNA is achieved by means of complementary short interfering RNA (siRNA) molecules. Short interfering RNA technology affords a potential tractable strategy to combat viral pathogenesis because siRNAs are specific, easy to design, and can be directed against multiple strains of a virus by targeting their conserved gene regions. In this review, we briefly summarize the current status of siRNA therapy for representative examples from different virus families. In addition, other aspects like their design, delivery, medical significance, bioinformatics resources, and limitations are also discussed.
Collapse
Affiliation(s)
- Abid Qureshi
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Vaqar Gani Tantray
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Altaf Rehman Kirmani
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Abdul Ghani Ahangar
- Biomedical Informatics Center, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| |
Collapse
|
19
|
Lau CML, Yu Y, Jahanmir G, Chau Y. Controlled release technology for anti-angiogenesis treatment of posterior eye diseases: Current status and challenges. Adv Drug Deliv Rev 2018; 126:145-161. [PMID: 29625138 DOI: 10.1016/j.addr.2018.03.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 12/27/2022]
Abstract
Antiangiogenic therapeutics, such as corticosteroids, VEGF targeting antibodies and aptamers have been demonstrated effective in controlling retinal and choroidal neovascularization related vision loss. However, to manage the chronic conditions, it requires long term and frequent intravitreal injections of these drugs, resulting in poor patient compliance and suboptimal treatment. In addition, emerging drugs such as tyrosine kinase inhibitors and siRNAs received much expectations, but the late stage clinical trials encountered various obstacles. Controlled release technology could improve the existing treatment regimen by extending therapeutic duration, reducing risks and burdens caused by frequent injections, and enabling new drugs to overcome the hurdles of translation. Here, we give qualitative and quantitative discussions about the principle mechanisms of polymeric reservoir, polymeric matrix and hydrogel systems. We also reveal the design rationales of the existing drug delivery and release systems in preclinical and clinical stages. Lastly, the animal models of ocular angiogenesis diseases are critically reviewed, which could help to facilitate the translation of controlled release technologies from bench to bedside.
Collapse
Affiliation(s)
- Chi Ming Laurence Lau
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong
| | - Yu Yu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong; Pleryon Therapeutics Limited, Hong Kong
| | - Ghodsiehsadat Jahanmir
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong
| | - Ying Chau
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong; The Hong Kong University of Science and Technology Shenzhen Institute, Shenzhen 518057, China.
| |
Collapse
|
20
|
Angelbello AJ, Chen JL, Childs-Disney JL, Zhang P, Wang ZF, Disney MD. Using Genome Sequence to Enable the Design of Medicines and Chemical Probes. Chem Rev 2018; 118:1599-1663. [PMID: 29322778 DOI: 10.1021/acs.chemrev.7b00504] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Rapid progress in genome sequencing technology has put us firmly into a postgenomic era. A key challenge in biomedical research is harnessing genome sequence to fulfill the promise of personalized medicine. This Review describes how genome sequencing has enabled the identification of disease-causing biomolecules and how these data have been converted into chemical probes of function, preclinical lead modalities, and ultimately U.S. Food and Drug Administration (FDA)-approved drugs. In particular, we focus on the use of oligonucleotide-based modalities to target disease-causing RNAs; small molecules that target DNA, RNA, or protein; the rational repurposing of known therapeutic modalities; and the advantages of pharmacogenetics. Lastly, we discuss the remaining challenges and opportunities in the direct utilization of genome sequence to enable design of medicines.
Collapse
Affiliation(s)
- Alicia J Angelbello
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jonathan L Chen
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jessica L Childs-Disney
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Peiyuan Zhang
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Zi-Fu Wang
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Matthew D Disney
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
21
|
Philp AM, Davis ET, Jones SW. Developing anti-inflammatory therapeutics for patients with osteoarthritis. Rheumatology (Oxford) 2017; 56:869-881. [PMID: 27498352 DOI: 10.1093/rheumatology/kew278] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Indexed: 12/30/2022] Open
Abstract
OA is the most common joint disorder in the world, but there are no approved therapeutics to prevent disease progression. Historically, OA has been considered a wear-and-tear joint disease, and efforts to identify and develop disease-modifying therapeutics have predominantly focused on direct inhibition of cartilage degeneration. However, there is now increasing evidence that inflammation is a key mediator of OA joint pathology, and also that the link between obesity and OA is not solely due to excessive load-bearing, suggesting therefore that targeting inflammation in OA could be a rewarding therapeutic strategy. In this review we therefore re-evaluate historical clinical trial data on anti-inflammatory therapeutics in OA patients, highlight some of the more promising emerging therapeutic targets and discuss the implications for future clinical trial design.
Collapse
Affiliation(s)
- Ashleigh M Philp
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, Medical School, Queen Elizabeth Hospital, University of Birmingham
| | - Edward T Davis
- The Royal Orthopaedic Hospital NHS Foundation Trust, Bristol Road South, Northfield, Birmingham
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, Medical School, Queen Elizabeth Hospital, University of Birmingham
| |
Collapse
|
22
|
Chernikov IV, Gladkikh DV, Meschaninova MI, Ven'yaminova AG, Zenkova MA, Vlassov VV, Chernolovskaya EL. Cholesterol-Containing Nuclease-Resistant siRNA Accumulates in Tumors in a Carrier-free Mode and Silences MDR1 Gene. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 6:209-220. [PMID: 28325287 PMCID: PMC5363506 DOI: 10.1016/j.omtn.2016.12.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/23/2016] [Accepted: 12/23/2016] [Indexed: 12/26/2022]
Abstract
Chemical modifications are an effective way to improve the therapeutic properties of small interfering RNAs (siRNAs), making them more resistant to degradation in serum and ensuring their delivery to target cells and tissues. Here, we studied the carrier-free biodistribution and biological activity of a nuclease-resistant anti-MDR1 cholesterol-siRNA conjugate in healthy and tumor-bearing severe combined immune deficiency (SCID) mice. The attachment of cholesterol to siRNA provided its efficient accumulation in the liver and in tumors, and reduced its retention in the kidneys after intravenous and intraperitoneal injection. The major part of cholesterol-siRNA after intramuscular and subcutaneous injections remained in the injection place. Confocal microscopy data demonstrated that cholesterol-siRNA spread deep in the tissue and was present in the cytoplasm of almost all the liver and tumor cells. The reduction of P-glycoprotein level in human KB-8-5 xenograft overexpressing the MDR1 gene by 60% was observed at days 5–6 after injection. Then, its initial level recovered by the eighth day. The data showed that, regardless of the mode of administration (intravenous, intraperitoneal, or peritumoral), cholesterol-siMDR efficiently reduced the P-glycoprotein level in tumors. The designed anti-MDR1 conjugate has potential as an adjuvant therapeutic for the reversal of multiple drug resistance of cancer cells.
Collapse
Affiliation(s)
- Ivan V Chernikov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia
| | - Daniil V Gladkikh
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia
| | - Mariya I Meschaninova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia
| | - Alya G Ven'yaminova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia
| | - Marina A Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia
| | - Valentin V Vlassov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia
| | - Elena L Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia.
| |
Collapse
|
23
|
Bi Y, Zhang Y, Cui C, Ren L, Jiang X. Gene-silencing effects of anti-survivin siRNA delivered by RGDV-functionalized nanodiamond carrier in the breast carcinoma cell line MCF-7. Int J Nanomedicine 2016; 11:5771-5787. [PMID: 27853365 PMCID: PMC5104303 DOI: 10.2147/ijn.s117611] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nanodiamond (ND) is a renowned material in nonviral small interfering RNA (siRNA) carrier field due to its unique physical, chemical, and biological properties. In our previous work, it was proven that ND could deliver siRNA into cells efficiently and downregulate the expression of desired protein. However, synthesizing a high-efficient tumor-targeting carrier using ND is still a challenge. In this study, a novel carrier, NDCONH(CH2)2NH-VDGR, was synthesized for siRNA delivery, and its properties were characterized with methods including Fourier transform infrared spectrometry, transmission electron microscopy, scanning electron microscopy, gel retardation assay, differential scanning calorimetry, confocal microscopy, releasing test, real-time polymerase chain reaction (PCR) assay, enzyme-linked immunosorbent assay (ELISA), flow cytometry, cytotoxicity assay, and gene-silencing efficacy assay in vitro and in vivo. The mechanism of NDCONH(CH2)2NH-VDGR/survivin-siRNA-induced tumor apoptosis was evaluated via flow cytometer assay using Annexin V–fluorescein isothiocyanate/propidium iodide staining method. The NDCONH(CH2)2NH-VDGR/survivin-siRNA nanoparticle with 60–110 nm diameter and 35.65±3.90 mV zeta potential was prepared. For real-time PCR assay, the results showed that the expression of survivin mRNA was reduced to 46.77%±6.3%. The expression of survivin protein was downregulated to 48.49%±2.25%, as evaluated by ELISA assay. MTT assay showed that NDCONH(CH2)2NH-VDGR/survivin-siRNA had an inhibitory effect on MCF-7 cell proliferation. According to these results, the survivin-siRNA could be delivered, transported, and released stably, which benefits in increasing the gene-silencing effect. Therefore, as an siRNA carrier, NDCONH(CH2)2NH-VDGR was suggested to be used in siRNA delivery system and in cancer treatments.
Collapse
Affiliation(s)
- Yanzhao Bi
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yifan Zhang
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Chunying Cui
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Lulu Ren
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xueyun Jiang
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
24
|
Park J, Park J, Pei Y, Xu J, Yeo Y. Pharmacokinetics and biodistribution of recently-developed siRNA nanomedicines. Adv Drug Deliv Rev 2016; 104:93-109. [PMID: 26686832 DOI: 10.1016/j.addr.2015.12.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/26/2015] [Accepted: 12/03/2015] [Indexed: 02/07/2023]
Abstract
Small interfering RNA (siRNA) is a promising drug candidate, expected to have broad therapeutic potentials toward various diseases including viral infections and cancer. With recent advances in bioconjugate chemistry and carrier technology, several siRNA-based drugs have advanced to clinical trials. However, most cases address local applications or diseases in the filtering organs, reflecting remaining challenges in systemic delivery of siRNA. The difficulty in siRNA delivery is in large part due to poor circulation stability and unfavorable pharmacokinetics and biodistribution profiles of siRNA. This review describes the pharmacokinetics and biodistribution of siRNA nanomedicines, focusing on those reported in the past 5years, and their pharmacological effects in selected disease models such as hepatocellular carcinoma, liver infections, and respiratory diseases. The examples discussed here will provide an insight into the current status of the art and unmet needs in siRNA delivery.
Collapse
|
25
|
Han H, Son S, Son S, Kim N, Yhee JY, Lee JH, Choi JS, Joo CK, Lee H, Lee D, Kim WJ, Kim SH, Kwon IC, Kim H, Kim K. Reducible Polyethylenimine Nanoparticles for Efficient siRNA Delivery in Corneal Neovascularization Therapy. Macromol Biosci 2016; 16:1583-1597. [DOI: 10.1002/mabi.201600051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 07/14/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Hyounkoo Han
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
- Department of Chemical and Biomolecular Engineering; Sogang University; Shinsu-dong Mapo-gu Seoul 121-742 South Korea
| | - Sohee Son
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
| | - Sejin Son
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
| | - Namho Kim
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
| | - Ji Young Yhee
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
| | - Jae Hyeop Lee
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
- Department of Chemical and Biomolecular Engineering; Sogang University; Shinsu-dong Mapo-gu Seoul 121-742 South Korea
| | - Jun-Sub Choi
- Department of Ophthalmology and Visual Science; Seoul St. Mary's Hospital; College of Medicine; The Catholic University of Korea; 505, Banpo-dong Seocho-gu Seoul 137-040 South Korea
| | - Choun-Ki Joo
- Department of Ophthalmology and Visual Science; Seoul St. Mary's Hospital; College of Medicine; The Catholic University of Korea; 505, Banpo-dong Seocho-gu Seoul 137-040 South Korea
| | - Hohyeon Lee
- Department of Chemical and Biomolecular Engineering; Sogang University; Shinsu-dong Mapo-gu Seoul 121-742 South Korea
| | - Duhwan Lee
- Center for Self-Assembly and Complexity; Institute for Basic Science (IBS); Pohang 790-784 South Korea
- Department of Chemistry Polymer Research Institute; Pohang University of Science and Technology (POSTECH); Pohang 790-784 South Korea
| | - Won Jong Kim
- Center for Self-Assembly and Complexity; Institute for Basic Science (IBS); Pohang 790-784 South Korea
- Department of Chemistry Polymer Research Institute; Pohang University of Science and Technology (POSTECH); Pohang 790-784 South Korea
| | - Sun Hwa Kim
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
| | - Ick Chan Kwon
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering; Sogang University; Shinsu-dong Mapo-gu Seoul 121-742 South Korea
| | - Kwangmeyung Kim
- Center for Theragnosis; Biomedical Research Institute Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 6 Seongbuk-gu Seoul 136-791 South Korea
| |
Collapse
|
26
|
Lee J, Ryoo NK, Han H, Hong HK, Park JY, Park SJ, Kim YK, Sim C, Kim K, Woo SJ, Park KH, Kim H. Anti-VEGF PolysiRNA Polyplex for the Treatment of Choroidal Neovascularization. Mol Pharm 2016; 13:1988-95. [DOI: 10.1021/acs.molpharmaceut.6b00148] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Jihwang Lee
- Department
of Chemical and Biomolecular Engineering, Sogang University, 35
Baekbeom-ro, Mapo-gu, Seoul 121-742, Republic of Korea
| | - Na-kyung Ryoo
- Department
of Ophthalmology, Seoul National University Bundang Hospital, 82,
Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
- Department
of Ophthalmology, Seoul National University College of Medicine, 71, Ihwajang-gil, Jongno-gu, Seoul, Korea
| | - Hyounkoo Han
- Department
of Chemical and Biomolecular Engineering, Sogang University, 35
Baekbeom-ro, Mapo-gu, Seoul 121-742, Republic of Korea
| | - Hye Kyoung Hong
- Department
of Ophthalmology, Seoul National University Bundang Hospital, 82,
Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
| | - Ji Yeon Park
- Department
of Ophthalmology, Seoul National University Bundang Hospital, 82,
Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
| | - Sang Jun Park
- Department
of Ophthalmology, Seoul National University Bundang Hospital, 82,
Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
- Department
of Ophthalmology, Seoul National University College of Medicine, 71, Ihwajang-gil, Jongno-gu, Seoul, Korea
| | - Yong-Kyu Kim
- Department
of Ophthalmology, Seoul National University Bundang Hospital, 82,
Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
- Department
of Ophthalmology, Seoul National University College of Medicine, 71, Ihwajang-gil, Jongno-gu, Seoul, Korea
| | - Changbeom Sim
- Department
of Chemical and Biomolecular Engineering, Sogang University, 35
Baekbeom-ro, Mapo-gu, Seoul 121-742, Republic of Korea
| | - Kwangmeyung Kim
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | - Se Joon Woo
- Department
of Ophthalmology, Seoul National University Bundang Hospital, 82,
Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
- Department
of Ophthalmology, Seoul National University College of Medicine, 71, Ihwajang-gil, Jongno-gu, Seoul, Korea
| | - Kyu Hyung Park
- Department
of Ophthalmology, Seoul National University Bundang Hospital, 82,
Gumi-ro 173 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
- Department
of Ophthalmology, Seoul National University College of Medicine, 71, Ihwajang-gil, Jongno-gu, Seoul, Korea
| | - Hyuncheol Kim
- Department
of Chemical and Biomolecular Engineering, Sogang University, 35
Baekbeom-ro, Mapo-gu, Seoul 121-742, Republic of Korea
| |
Collapse
|
27
|
Zhou Y, Zhou G, Tian C, Jiang W, Jin L, Zhang C, Chen X. Exosome-mediated small RNA delivery for gene therapy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 7:758-771. [DOI: 10.1002/wrna.1363] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Yu Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS); School of Life Sciences, Nanjing University; Nanjing China
| | - Geyu Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS); School of Life Sciences, Nanjing University; Nanjing China
| | - Chenfei Tian
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS); School of Life Sciences, Nanjing University; Nanjing China
| | - Waner Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS); School of Life Sciences, Nanjing University; Nanjing China
| | - Ling Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS); School of Life Sciences, Nanjing University; Nanjing China
| | - Chenyu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS); School of Life Sciences, Nanjing University; Nanjing China
| | - Xi Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences (NAILS); School of Life Sciences, Nanjing University; Nanjing China
| |
Collapse
|
28
|
The exit strategy: Pharmacological modulation of extracellular matrix production and deposition for better aqueous humor drainage. Eur J Pharmacol 2016; 787:32-42. [PMID: 27112663 DOI: 10.1016/j.ejphar.2016.04.048] [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] [Received: 01/27/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 01/28/2023]
Abstract
Primary open angle glaucoma (POAG) is an optic neuropathy and an irreversible blinding disease. The etiology of glaucoma is not known but numerous risk factors are associated with this disease including aging, elevated intraocular pressure (IOP), race, myopia, family history and use of steroids. In POAG, the resistance to the aqueous humor drainage is increased leading to elevated IOP. Lowering the resistance and ultimately the IOP has been the only way to slow disease progression and prevent vision loss. The primary drainage pathway comprising of the trabecular meshwork (TM) is made up of relatively large porous beams surrounded by extracellular matrix (ECM). Its juxtacanalicular tissue (JCT) or the cribriform meshwork is made up of cells embedded in dense ECM. The JCT is considered to offer the major resistance to the aqueous humor outflow. This layer is adjacent to the endothelial cells forming Schlemm's canal, which provides approximately 10% of the outflow resistance. The ECM in the TM and the JCT undergoes continual remodeling to maintain normal resistance to aqueous humor outflow. It is believed that the TM is a major contributor of ECM proteins and evidence points towards increased ECM deposition in the outflow pathway in POAG. It is not clear how and from where the ECM components emerge to hinder the normal aqueous humor drainage. This review focuses on the involvement of the ECM in ocular hypertension and glaucoma and the mechanisms by which various ocular hypotensive drugs, both current and emerging, target ECM production, remodeling, and deposition.
Collapse
|
29
|
Bobbin ML, Rossi JJ. RNA Interference (RNAi)-Based Therapeutics: Delivering on the Promise? Annu Rev Pharmacol Toxicol 2016; 56:103-22. [DOI: 10.1146/annurev-pharmtox-010715-103633] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Maggie L. Bobbin
- Molecular Pathology Unit and
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, Massachusetts 02129;
| | - John J. Rossi
- Beckman Research Institute, City of Hope, Duarte, California 91010;
| |
Collapse
|
30
|
Aljuffali IA, Lin YK, Fang JY. Noninvasive approach for enhancing small interfering RNA delivery percutaneously. Expert Opin Drug Deliv 2015; 13:265-80. [DOI: 10.1517/17425247.2016.1121988] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
31
|
Zarembinski TI, Doty NJ, Erickson IE, Srinivas R, Wirostko BM, Tew WP. Thiolated hyaluronan-based hydrogels crosslinked using oxidized glutathione: an injectable matrix designed for ophthalmic applications. Acta Biomater 2014; 10:94-103. [PMID: 24096152 DOI: 10.1016/j.actbio.2013.09.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 08/10/2013] [Accepted: 09/23/2013] [Indexed: 02/08/2023]
Abstract
Future ophthalmic therapeutics will require the sustained delivery of bioactive proteins and nucleic acid-based macromolecules and/or provide a suitable microenvironment for the localization and sustenance of reparative progenitor cells after transplantation into or onto the eye. Water-rich hydrogels are ideal vehicles for such cargo, but few have all the qualities desired for novel ophthalmic use, namely in situ gelation speed, cytocompatibility, biocompatibility and capacity to functionalize. We describe here the development of an ophthalmic-compatible crosslinking system using oxidized glutathione (GSSG), a physiologically relevant molecule with a history of safe use in humans. When GSSG is used in conjunction with an existing hyaluronate-based, in situ crosslinkable hydrogel platform, gels form in less than 5 min using the thiol-disulfide exchange reaction. This GSSG hydrogel supports the 3-D culture of adipose-derived stem cells in vitro and shows biocompatibility in preliminary intracutaneous and subconjunctival experiments in vivo. In addition, the thiol-disulfide exchange reaction can also be used in conjunction with other thiol-compatible chemistries to covalently link peptides for more complex formulations. These data suggest that this hydrogel could be well suited for local ocular delivery, focusing initially on front of the eye therapies. Subsequent uses of the hydrogel include delivery of back of the eye treatments and eventually into other soft, hyaluronan-rich tissues such as those from the liver and brain.
Collapse
|