1
|
Silverstein SM, Oddone F, Kolko M, Brinkmann CK, Christie WC, Bicket AK, Gous PNJ, Luebke J, Maram J, Nguyen A, Craven ER, Pu Y, Jiao J, Bejanian M, Robinson MR. Safety and Longevity of Intraocular Pressure Control After Bimatoprost Implant Administration: Interim Analysis of a Phase 3b Clinical Trial (TRITON). Drugs 2025; 85:557-570. [PMID: 39985740 PMCID: PMC11947067 DOI: 10.1007/s40265-025-02154-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Bimatoprost implant 10 µg is an intracameral, biodegradable implant that slowly releases bimatoprost to lower intraocular pressure (IOP). This study was designed to evaluate safety and the duration of the IOP-lowering effect after single and as-needed repeat administration of the bimatoprost implant in patients with open-angle glaucoma (OAG) and ocular hypertension (OHT). PATIENTS AND METHODS This study is an interim analysis of an ongoing, prospective, open-label, multicenter study in patients with OAG or OHT who are inadequately managed with topical IOP-lowering medication for reasons other than efficacy. IOP-lowering rescue treatment is allowed if implant retreatment criteria are not met. The primary endpoint is time to retreatment/rescue after the initial implant administration analyzed with the Kaplan-Meier method. Key safety measures include treatment-emergent adverse events (TEAEs) and reading-center evaluation of central corneal endothelial cell density (CECD). Analysis of data collected through 15 September 2023 focused on outcomes after a single or two implants. RESULTS In total, 441 patients received the 10-µg bimatoprost implant in the study eye on day 1 (cycle 1), 179 patients received a second administration (cycle 2), and 378 patients had at least 12 months of follow-up data available. The median time (95% confidence interval) from the first administration to a second administration or rescue was 392 (369, 485) days; the probability of not requiring retreatment or rescue by day 360 was 57.5%. A second implant administration similarly provided a long duration of IOP control. The baseline mean (standard error, SE) IOP was 25.6 (0.14) mmHg; the mean (SE) change from baseline IOP in unrescued eyes after a single administration was - 7.5 (0.21) mmHg at week 24 and - 6.4 (0.28) mmHg at month 12. Conjunctival hyperemia, typically associated with the administration procedure, was the most common ocular TEAE (cycle 1, 14.3%; cycle 2, 12.8%). Mean (SE) percentage change in CECD from baseline at 12 months after administration was - 4.3 (0.81)% in cycle 1 and - 8.5 (2.22)% in cycle 2. The cycle 1 implant was no longer visible or ≤ 25% of initial size in 66.3% and 94.3% of study eyes at months 12 and 24, respectively. CONCLUSIONS In this interim analysis based on available data, the IOP-lowering effect of the initial administration of the 10-µg bimatoprost implant was well maintained for > 1 year in most patients. Results after a second administration were comparable. The safety profile of initial and repeat administration was acceptable. TRIAL REGISTRY ClinicalTrials.gov identifier NCT03850782; registered 20 February 2019.
Collapse
Affiliation(s)
- Steven M Silverstein
- Silverstein Eye Centers, 4240 Blue Ridge Blvd. Suite 1000, Kansas City, MO, 64133, USA.
| | | | - Miriam Kolko
- Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Amanda K Bicket
- Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Jan Luebke
- Eye Center, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | | | - Yongjia Pu
- Allergan, an Abbvie company, Irvine, CA, USA
| | - Jenny Jiao
- Allergan, an Abbvie company, Irvine, CA, USA
| | | | | |
Collapse
|
2
|
Tan SS, Wong TT. Drug delivery systems in glaucoma - Current innovations and future perspectives. Asia Pac J Ophthalmol (Phila) 2025; 14:100163. [PMID: 40010440 DOI: 10.1016/j.apjo.2025.100163] [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: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/28/2025] Open
Abstract
Glaucoma is the leading cause of irreversible visual loss worldwide, and as yet, there is no cure. The only evidence-based treatment to slow progression is by lowering intraocular pressure (IOP). Despite the development of new topical medications to reduce IOP, the major limitation of eyedrops lies in human and anatomical factors, namely patient compliance and poor bioavailability, making current medical glaucoma treatment ineffective. In this manuscript, we summarise the limitations of traditional topical anti-glaucoma therapy and study current drug delivery systems to lower IOP, with focus on the only two that have made FDA-approval- Durysta and iDose TR. We highlight their limitations and discuss real-world economic challenges that make it prohibitively difficult for these drug delivery systems to be more widely adopted in daily practice. In this perspective, we also introduce gene therapy as a novel therapeutic option to target downstream pathways of IOP regulation, neuroprotection of the optic nerve, and reducing mitochondrial stress to delay the progression of glaucoma. We discuss promising results of gene therapy for glaucoma treatment in in vivo animal models as well. We also explore the concept of novel nanoparticle-based drug delivery systems, which have the advantage of being highly modifiable and customisable, able to incorporate large amounts of cargo while maintaining a high transfection efficacy, and at a fraction of the cost. Lastly, we propose that nanomedicine, in conjunction with gene therapy, offers a promising solution to the aforementioned challenges of current glaucoma therapy, and can herald a new era of sustained glaucoma treatment.
Collapse
Affiliation(s)
- Shayne S Tan
- Singapore National Eye Centre, Singapore, Singapore Eye Research Institute, Singapore
| | - Tina T Wong
- Singapore National Eye Centre, Singapore, Singapore Eye Research Institute, Singapore.
| |
Collapse
|
3
|
Jiao C, Prabakaran G, Berk T, Hossain HA, Arepalli S. Persistent bilateral sclerouveitis following bimatoprost implantation and removal. Am J Ophthalmol Case Rep 2025; 37:102240. [PMID: 39839914 PMCID: PMC11750282 DOI: 10.1016/j.ajoc.2024.102240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Purpose To report a case of persistent bilateral sclerouveitis following bilateral bimatoprost implantation (Durysta) that required implant removal and oral steroid course. Observation A 75-year-old Caucasian male with no prior ocular inflammation experienced bilateral sclerouveitis post bilateral bimatoprost implantation. Despite implant removal from both eyes, the ocular inflammation persisted, showing cystoid macular edema in both eyes and exudative retinal detachment in the right. A comprehensive assessment revealed no clear etiology and no definitive systematic inflammatory syndrome. The ocular inflammation was unresponsive to topical steroids and only receded following weeks-long course of oral steroids. Eventually, the patient required immunosuppression due to recurrence of the inflammation. Conclusion and importance The persistence and severity of this inflammatory response to bimatoprost implants, despite its removal, highlight the importance of considering patient-specific risk factors and tailoring management accordingly. Clinicians should be prepared for possible severe reactions requiring intervention.
Collapse
Affiliation(s)
- Cheng Jiao
- Medical College of Georgia, Augusta University, Augusta, GA, USA
- University of North Carolina, Chapel Hill, NC, USA
| | | | - Thomas Berk
- Emory Eye Center, Emory University, Atlanta, GA, USA
| | | | | |
Collapse
|
4
|
Cheng CM, Rehmani C, Chin J. Prostaglandin Intracameral Implants for Ocular Hypertension and Open-Angle Glaucoma. J Pharm Technol 2025:87551225251313707. [PMID: 39911138 PMCID: PMC11791955 DOI: 10.1177/87551225251313707] [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: 02/07/2025] Open
Abstract
Background: Sustained-release prostaglandin intracameral implants are new targeted treatment options for open-angle glaucoma or ocular hypertension that lower intraocular pressure (IOP) and reduce or eliminate the need for topical eye drops. Objective: To summarize evidence supporting prostaglandin intracameral implants for treatment of ocular hypertension or open angle glaucoma and identify patient populations most likely to benefit from these treatments. Data sources: A PubMed search (1/1/2016 to 10/1/2024) was conducted to identify randomized, controlled clinical trials for bimatoprost 10-μg and travoprost 75-μg intracameral implants. Manufacturer prescribing information, formulary dossiers, Food and Drug Administration (FDA) clinical reviews and glaucoma clinical treatment guidelines were also reviewed. Study selection and data extraction: English-language randomized controlled trials involving bimatoprost 10-μg or travoprost 75-μg intracameral implants were included. Data synthesis: Bimatoprost and travoprost intracameral implants demonstrated noninferior IOP reduction compared to timolol eye drops in phase 3 trials, with sustained effects up to 12 and 36 months, respectively. The FDA-approved implants are limited to a single administration to the affected eye to minimize corneal risks. The travoprost implant contains a titanium reservoir and requires surgical placement, while the bimatoprost implant is biodegradable and can be placed in a clinic setting. There are no studies directly comparing the safety and efficacy of the two intracameral implants. Conclusions: Prostaglandin intracameral implants are a novel approach to reducing medication burden while delivering sustained IOP reducing effects. Pharmacists should be aware of efficacy and safety considerations of these implants relative to available topical treatments for ocular hypertension or open angle glaucoma.
Collapse
Affiliation(s)
| | | | - Jordan Chin
- School of Pharmacy, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
5
|
Vagiakis I, Papadopoulou EP, Amaxilati E, Tsiropoulos GN, Konstas AG, Panos GD. Bimatoprost Intracameral Implant (Durysta®): A New Era in Glaucoma Management Through Sustained-Release Innovation. Drug Des Devel Ther 2025; 19:703-714. [PMID: 39906697 PMCID: PMC11792881 DOI: 10.2147/dddt.s506520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
The bimatoprost intracameral implant (Durysta®) offers a sustained-release approach to glaucoma management, providing consistent intraocular pressure (IOP) reduction over several months and reducing the need for daily topical therapies. This review evaluates its pharmacology, efficacy, and safety, using data from pivotal clinical trials and recent real-world studies. The implant achieves IOP reductions comparable to topical prostaglandin analogs, with benefits for patient adherence and fewer common side effects. However, repeat administrations are associated with adverse effects such as endothelial cell loss, highlighting the need for optimized re-dosing schedules. Future research should explore its use in advanced glaucomas, cost-effectiveness, and combination with other IOP-lowering treatments. The bimatoprost intracameral implant represents a promising innovation in glaucoma therapy with potential for improved patient outcomes.
Collapse
Affiliation(s)
- Iordanis Vagiakis
- First Department of Ophthalmology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni P Papadopoulou
- First Department of Ophthalmology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efstratia Amaxilati
- First Department of Ophthalmology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios N Tsiropoulos
- First Department of Ophthalmology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasios G Konstas
- First Department of Ophthalmology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios D Panos
- First Department of Ophthalmology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Division of Ophthalmology and Visual Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
6
|
de Sousa Moreira A, Lopes B, Sousa AC, Coelho A, Sousa P, Araújo A, Delgado E, Alvites R, Maurício AC. Stem Cell-Based Therapies for Glaucoma Treatment: A Review Bridging the Gap in Veterinary Patients. Int J Mol Sci 2024; 26:232. [PMID: 39796087 PMCID: PMC11719664 DOI: 10.3390/ijms26010232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Retinal diseases are characterized by progressive damage to retinal cells, leading to irreversible vision loss. Among these, glaucoma stands out as a multifactorial neurodegenerative disease involving elevated intraocular pressure, retinal ganglion cell apoptosis, and optic nerve damage, ultimately resulting in blindness in both humans and dogs. Stem cell-based therapies have emerged as a promising therapeutic option for such conditions due to their regenerative and neuroprotective potential. These therapies, particularly those based on mesenchymal stem cells, offer the potential to repair and protect retinal tissues through the bioactive molecules (growth factors, cytokines, chemokines) secreted, their secretome. However, research in this field, especially on the use of umbilical cord mesenchymal stem cells' secretome, remains sparse. Most clinical trials focus on human glaucomatous patients, leaving a significant gap in veterinary patients' application, especially in dogs, with additional research being needed to determine its usefulness in canine glaucoma treatment. Future studies should aim to evaluate these therapies across both human and veterinary contexts, broadening treatment possibilities for glaucoma.
Collapse
Affiliation(s)
- Alícia de Sousa Moreira
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - André Coelho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Ana Araújo
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Esmeralda Delgado
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
- Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
- Instituto Universitário de Ciências da Saúde (CESPU), Avenida Central de Gandra n° 1317, 4585-116 Paredes, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| |
Collapse
|
7
|
Salvetat ML, Toro MD, Pellegrini F, Scollo P, Malaguarnera R, Musa M, Mereu L, Tognetto D, Gagliano C, Zeppieri M. Advancing Glaucoma Treatment During Pregnancy and Breastfeeding: Contemporary Management Strategies and Prospective Therapeutic Developments. Biomedicines 2024; 12:2685. [PMID: 39767592 PMCID: PMC11727364 DOI: 10.3390/biomedicines12122685] [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: 10/22/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/15/2025] Open
Abstract
The management of glaucoma in pregnancy and breastfeeding requires a careful evaluation of treatment choices to guarantee the well-being of both the mother and the developing fetus. This review explores the intricacies of controlling glaucoma in pregnant and breastfeeding women, including a comprehensive overview of existing glaucoma treatment methods, clinical guidelines, and future therapeutic approaches. The efficacy and safety profiles of traditional treatment approaches, such as topical and systemic medicines and surgical treatments, are evaluated specifically about their use during pregnancy and breastfeeding. The significance of personalized treatment programs to achieve a balance between controlling intraocular pressure and ensuring the safety of the fetus and the newborn and the importance of a multidisciplinary approach that includes ophthalmologists, obstetricians, and other healthcare experts are underlined. Non-pharmacological therapies, lifestyle adjustments, and the importance of patient education in the management of glaucoma during pregnancy and the post-partum period are also examined. Advancing our comprehension of and strategy toward glaucoma can reduce the effects of glaucoma on maternal, fetal, and newborn well-being.
Collapse
Affiliation(s)
- Maria Letizia Salvetat
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, Via Montereale 24, 33170 Pordenone, Italy;
| | - Mario Damiano Toro
- Eye Clinic, Public Health Department, Federico II University, Via Pansini 5, 80131 Naples, Italy
- Department of Special Surgery, University of Jordan, Queen Rania St, Amman 11942, Jordan
| | - Francesco Pellegrini
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, Via Montereale 24, 33170 Pordenone, Italy;
| | - Paolo Scollo
- Department of Medicine and Surgery, University of Enna “Kore”, Piazza dell’Università, 94100 Enna, Italy
- Gynecology and Obstetrics Unit, Cannizzaro Hospital, 95126 Catania, Italy
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, University of Enna “Kore”, Piazza dell’Università, 94100 Enna, Italy
| | - Mutali Musa
- Department of Optometry, University of Benin, Benin City 300238, Edo State, Nigeria
| | - Liliana Mereu
- Gynecology and Obstetrics Unit, Policlinico G. Rodolico, Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95124 Catania, Italy
| | - Daniele Tognetto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, Piazza dell’Università, 94100 Enna, Italy
- Mediterranean Foundation “G.B. Morgagni”, 95125 Catania, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
8
|
Pandey PK, Jain M, Jha PK. Drug delivery from a ring implant attached to intraocular lens: An in-silico investigation. J Pharm Sci 2024; 113:3332-3343. [PMID: 39245324 DOI: 10.1016/j.xphs.2024.09.001] [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: 06/09/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/10/2024]
Abstract
Multiple iterations required to design ocular implants, which will last for the desired operational period of months or even years, necessitate the use of in-silico models for ocular drug delivery. In this study, we developed an in-silico model to simulate the flow of Aqueous Humor (AH) and drug delivery from an implant to the Trabecular Meshwork (TM). The implant, attached to the side of the intraocular lens (IOL), and the TM are treated as porous media, with their effects on AH flow accounted for using the Darcy equation. This model accurately predicts the physiological values of Intraocular Pressure (IOP) for both healthy individuals and glaucoma patients, as reported in the literature. Results reveal that the effective diffusivity of the drug within the implant is the critical parameter that can alter the bioavailability time period (BTP) from a few days to months. Intuitively, BTP should increase as effective diffusivity decreases. However, we discovered that with lower levels of initial drug loading, BTP declines when effective diffusivity falls below a specific threshold. Our findings further reveal that, while AH flow has a minimal effect on the drug release profile at the implant site, it significantly impacts drug availability at the TM.
Collapse
Affiliation(s)
- Pawan Kumar Pandey
- Department of Chemical Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | | | - Prateek K Jha
- Department of Chemical Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
9
|
Elhusseiny AM, Aref AA. Sustained Release Therapies with the Prostaglandin Analogues Intracameral Implants. Ophthalmol Ther 2024; 13:1833-1839. [PMID: 38761359 PMCID: PMC11178720 DOI: 10.1007/s40123-024-00965-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/01/2024] [Indexed: 05/20/2024] Open
Abstract
The travoprost intracameral implant was recently approved by the US Food and Drug Administration for sustained release medical treatment of open-angle glaucoma in the USA. The approval represents a substantial and progressive step forward in the area of sustained-release glaucoma therapy. Topical intraocular pressure-lowering medications for the treatment of glaucoma are faced with a host of challenges for long-term and usually lifelong care. A changing paradigm in glaucoma management involves first-line interventions with laser modalities, micro-invasive surgeries, and sustained-release treatment platforms. Future needs in the area of sustained-release therapy include a non-prostaglandin drug delivery platform and longer-term treatments that do not require surgical reintervention.
Collapse
Affiliation(s)
- Abdelrahman M Elhusseiny
- Department of Ophthalmology, Harvey and Bernice Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ahmad A Aref
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, 1009 S Wood St, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
Choi EY, Johnson NA, Stinnett S, Rosdahl J, Moya F, Herndon LW. The Effect of Bimatoprost Implant on Glaucoma Patients: An Observational Study. J Glaucoma 2024; 33:431-436. [PMID: 38506759 DOI: 10.1097/ijg.0000000000002368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/20/2024] [Indexed: 03/21/2024]
Abstract
PRCIS In this retrospective study of glaucoma patients receiving the bimatoprost implant at Duke Eye Center, the number of topical intraocular pressure-lowering medications was significantly reduced through 12 months after the implant. PURPOSE To study the effects of the bimatoprost implant on intraocular pressure (IOP) and the need for topical IOP-lowering medications in glaucoma patients in the clinical practice setting. PATIENTS AND METHODS Patients who received the bimatoprost implant at Duke Eye Center from November 2020 to October 2021 were identified. Exclusion criteria included addition of other IOP-lowering medications concurrent with the implant and <1 month of follow-up. The change in IOP and number of topical IOP-lowering medications from baseline to months 1, 3, 6, 9, and 12 after the implant was calculated. Subgroup analysis was performed for different glaucoma severities. RESULTS A total of 63 patients and 92 eyes were included (mean age 77.8 ± 10.1 years). Glaucoma severity ranged from mild (11%), moderate (30%), to severe (54%). There was a nonsignificant decrease in IOP at all timepoints. The mean number of topical IOP-lowering medications significantly decreased by 0.81, 0.75, 0.63, 0.70, and 0.67 at month 1, 3, 6, 9, and 12, respectively (all P < 0.001). There was no significant change in the total number of medications, including the bimatoprost implant. When divided by glaucoma severity, the reduction in the number of topical medications was significant at 1, 3, and 6 months for mild/moderate disease and at 1 month for severe disease. During the follow-up period, 19 eyes underwent additional laser or surgical procedures, 68% of which had a history of prior incisional glaucoma surgery. CONCLUSIONS The bimatoprost implant may reduce the need for topical IOP-lowering agents over a 1-year period, especially in mild to moderate-stage glaucoma. The efficacy of the implant may be more limited in severe glaucoma, and further work is needed to characterize its long-term effects.
Collapse
Affiliation(s)
- Eun Young Choi
- Department of Ophthalmology, Duke University, Durham, NC
| | | | | | | | | | | |
Collapse
|
11
|
Zeppieri M, Gagliano C, Spadea L, Salati C, Chukwuyem EC, Enaholo ES, D’Esposito F, Musa M. From Eye Care to Hair Growth: Bimatoprost. Pharmaceuticals (Basel) 2024; 17:561. [PMID: 38794131 PMCID: PMC11124470 DOI: 10.3390/ph17050561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Bimatoprost has emerged as a significant medication in the field of medicine over the past several decades, with diverse applications in ophthalmology, dermatology, and beyond. Originally developed as an ocular hypotensive agent, it has proven highly effective in treating glaucoma and ocular hypertension. Its ability to reduce intraocular pressure has established it as a first-line treatment option, improving management and preventing vision loss. In dermatology, bimatoprost has shown promising results in the promotion of hair growth, particularly in the treatment of alopecia and hypotrichosis. Its mechanism of action, stimulating the hair cycle and prolonging the growth phase, has led to the development of bimatoprost-containing solutions for enhancing eyelash growth. AIM The aim of our review is to provide a brief description, overview, and studies in the current literature regarding the versatile clinical use of bimatoprost in recent years. This can help clinicians determine the most suitable individualized therapy to meet the needs of each patient. METHODS Our methods involve a comprehensive review of the latest advancements reported in the literature in bimatoprost formulations, which range from traditional eye drops to sustained-release implants. These innovations offer extended drug delivery, enhance patient compliance, and minimize side effects. RESULTS The vast literature published on PubMed has confirmed the clinical usefulness of bimatoprost in lowering intraocular pressure and in managing patients with glaucoma. Numerous studies have shown promising results in dermatology and esthetics in promoting hair growth, particularly in treating alopecia and hypotrichosis. Its mechanism of action involves stimulating the hair cycle and prolonging the growth phase, leading to the development of solutions that enhance eyelash growth. The global use of bimatoprost has expanded significantly, with applications growing beyond its initial indications. Ongoing research is exploring its potential in glaucoma surgery, neuroprotection, and cosmetic procedures. CONCLUSIONS Bimatoprost has shown immense potential for addressing a wide range of therapeutic needs through various formulations and advancements. Promising future perspectives include the exploration of novel delivery systems such as contact lenses and microneedles to further enhance drug efficacy and patient comfort. Ongoing research and future perspectives continue to shape its role in medicine, promising further advancements and improved patient outcomes.
Collapse
Affiliation(s)
- Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, Piazza dell’Università, 94100 Enna, Italy
- Eye Clinic, Catania University, San Marco Hospital, Viale Carlo Azeglio Ciampi, 95121 Catania, Italy
| | - Leopoldo Spadea
- Eye Clinic, Policlinico Umberto I, “ Sapienza” University of Rome, 00142 Rome, Italy
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | | | | | - Fabiana D’Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, London NW1 5QH, UK
| | - Mutali Musa
- Department of Optometry, University of Benin, Benin City 300238, Nigeria;
| |
Collapse
|
12
|
Bowers ME, Wong MK, Ventimiglia J, Nicknam RM, Moster MR, Pro MJ, Dale E, Kolomeyer NN, Lee D, Zheng CX. Effect of bimatoprost sustained-release intracameral implant on intraocular pressure and medication burden in patients with prior glaucoma surgery. J Fr Ophtalmol 2024; 47:103996. [PMID: 37926661 DOI: 10.1016/j.jfo.2023.07.016] [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: 05/15/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 11/07/2023]
Abstract
The present retrospective study evaluated intraocular pressure (IOP) and medication burden after bimatoprost sustained-release (bimatoprost SR, Durysta, Allergan) implantation in patients with glaucoma. A secondary objective was to examine an effect of bimatoprost SR in a subset of patients with prior minimally invasive and incisional glaucoma surgery. A retrospective chart review of 122 eyes that received bimatoprost SR by 6 glaucoma specialists at Wills Eye Hospital between March 2020 and September 2021 was performed. One hundred and eighteen eyes from 84 patients had a reduction in IOP (18.5±5.7mmHg vs. 16.0±5.4mmHg, P<0.01) and required fewer glaucoma medications (2.1±1.4 vs. 1.2±1.2, P<0.01) after bimatoprost SR implantation. In 41 eyes from 31 patients who previously underwent glaucoma surgery (including iStent, goniotomy, trabeculectomy, Xen Gel Stent, or tube shunt surgery), medication burden was decreased after bimatoprost SR implantation (1.9±1.3 vs. 1.0±1.0, P<0.001). These data suggest that bimatoprost SR is an efficacious treatment modality for glaucoma, even in post-surgical patients.
Collapse
Affiliation(s)
- M E Bowers
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States.
| | - M K Wong
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States
| | - J Ventimiglia
- University of Maryland at College Park, 20742 College Park, MD, United States
| | - R M Nicknam
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States; Glaucoma Service, Wills Eye Hospital, 840, Walnut Street, Suite 1140, 19107 Philadelphia, PA, United States
| | - M R Moster
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States; Glaucoma Service, Wills Eye Hospital, 840, Walnut Street, Suite 1140, 19107 Philadelphia, PA, United States
| | - M J Pro
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States; Glaucoma Service, Wills Eye Hospital, 840, Walnut Street, Suite 1140, 19107 Philadelphia, PA, United States
| | - E Dale
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States; Glaucoma Service, Wills Eye Hospital, 840, Walnut Street, Suite 1140, 19107 Philadelphia, PA, United States
| | - N N Kolomeyer
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States; Glaucoma Service, Wills Eye Hospital, 840, Walnut Street, Suite 1140, 19107 Philadelphia, PA, United States
| | - D Lee
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States; Glaucoma Service, Wills Eye Hospital, 840, Walnut Street, Suite 1140, 19107 Philadelphia, PA, United States
| | - C X Zheng
- Sidney-Kimmel Medical College, Thomas Jefferson University, 1025, Walnut Street, Suite 100, 19107 Philadelphia, PA, United States; Glaucoma Service, Wills Eye Hospital, 840, Walnut Street, Suite 1140, 19107 Philadelphia, PA, United States
| |
Collapse
|
13
|
Bedrood S, Berdahl J, Sheybani A, Singh IP. Alternatives to Topical Glaucoma Medication for Glaucoma Management. Clin Ophthalmol 2023; 17:3899-3913. [PMID: 38111854 PMCID: PMC10726774 DOI: 10.2147/opth.s439457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023] Open
Abstract
Topical glaucoma medications have favorable safety and efficacy, but their use is limited by factors such as side effects, nonadherence, costs, ocular surface disease, intraocular pressure fluctuations, diminished quality of life, and the inherent difficulty of penetrating the corneal surface. Although traditionally these limitations have been accepted as an inevitable part of glaucoma treatment, a rapidly-evolving arena of minimally invasive surgical and laser interventions has initiated the beginnings of a reevaluation of the glaucoma treatment paradigm. This reevaluation encompasses an overall shift away from the reactive, topical-medication-first default and a shift toward earlier intervention with laser or surgical therapies such as selective laser trabeculoplasty, sustained-release drug delivery, and micro-invasive glaucoma surgery. Aside from favorable safety, these interventions may have clinically important attributes such as consistent IOP control, cost-effectiveness, independence from patient adherence, prevention of disease progression, and improved quality of life.
Collapse
Affiliation(s)
| | | | - Arsham Sheybani
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
14
|
Wu CM. New Glaucoma Medications. Int Ophthalmol Clin 2023; 63:13-21. [PMID: 37755441 DOI: 10.1097/iio.0000000000000482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
|
15
|
Quaranta L, Novella A, Tettamanti M, Pasina L, Weinreb RN, Nobili A. Adherence and Persistence to Medical Therapy in Glaucoma: An Overview. Ophthalmol Ther 2023; 12:2227-2240. [PMID: 37311908 PMCID: PMC10441906 DOI: 10.1007/s40123-023-00730-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/28/2023] [Indexed: 06/15/2023] Open
Abstract
Glaucoma is a group of progressive optic neuropathies characterized by loss of retinal ganglion cells and visual field deterioration. Despite the fact that the underlying pathophysiology of glaucoma remains unknown, elevated intraocular pressure (IOP) is a well-established risk factor, and the only factor that can be modified. Robust evidence from epidemiological studies and clinical trials has clearly demonstrated the benefits of IOP control in reducing the risk of glaucoma progression. IOP-lowering therapy by the means of eye drops remains a first-line treatment option. However, like other chronic and asymptomatic conditions, many patients with glaucoma have difficulties in maintaining high rates of adherence persistence to prescribed medications. On average, patients with chronic medical conditions take 30-70% of the prescribed medication doses, and on average 50% discontinue medications in the first months of therapy. The ophthalmic literature shows similarly low rates of adherence to treatment. Indeed, poor adherence is associated with disease progression and increased complication rates, as well as healthcare costs. The present review analyzes and discusses the causes of variability of the adherence to the prescribed drugs. The education of patients about glaucoma and the potential consequences of insufficient adherence and persistence seems fundamental to maximize the probability of treatment success and therefore prevent visual disability to avoid unnecessary healthcare costs.
Collapse
Affiliation(s)
| | - Alessio Novella
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mauro Tettamanti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Pasina
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Robert N Weinreb
- Viterbi Family Department of Ophthalmology, and Shiley Eye Institute, Hamilton Glaucoma Center, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
16
|
Satyanarayana SD, Abu Lila AS, Moin A, Moglad EH, Khafagy ES, Alotaibi HF, Obaidullah AJ, Charyulu RN. Ocular Delivery of Bimatoprost-Loaded Solid Lipid Nanoparticles for Effective Management of Glaucoma. Pharmaceuticals (Basel) 2023; 16:1001. [PMID: 37513913 PMCID: PMC10385266 DOI: 10.3390/ph16071001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Glaucoma is a progressive optic neuropathy characterized by a rise in the intraocular pressure (IOP) leading to optic nerve damage. Bimatoprost is a prostaglandin analogue used to reduce the elevated IOP in patients with glaucoma. The currently available dosage forms for Bimatoprost suffer from relatively low ocular bioavailability. The objective of this study was to fabricate and optimize solid lipid nanoparticles (SLNs) containing Bimatoprost for ocular administration for the management of glaucoma. Bimatoprost-loaded SLNs were fabricated by solvent evaporation/ultrasonication technique. Glyceryl Monostearate (GMS) was adopted as solid lipid and poloxamer 407 as surfactant. Optimization of SLNs was conducted by central composite design. The optimized formulation was assessed for average particle size, entrapment efficiency (%), zeta potential, surface morphology, drug release study, sterility test, isotonicity test, Hen's egg test-chorioallantoic membrane (HET-CAM) test and histopathology studies. The optimized Bimatoprost-loaded SLNs formulation had an average size of 183.3 ± 13.3 nm, zeta potential of -9.96 ± 1.2 mV, and encapsulation efficiency percentage of 71.8 ± 1.1%. Transmission electron microscopy (TEM) study revealed the nearly smooth surface of formulated particles with a nano-scale size range. In addition, SLNs significantly sustained Bimatoprost release for up to 12 h, compared to free drug (p < 005). Most importantly, HET-CAM test nullified the irritancy of the formulation was verified its tolerability upon ocular use, as manifested by a significant reduction in mean irritation score, compared to positive control (1% sodium dodecyl sulfate; p < 0.001). Histopathology study inferred the absence of any signs of cornea tissue damage upon treatment with Bimatoprost optimized formulation. Collectively, it was concluded that SLNs might represent a viable vehicle for enhancing the corneal permeation and ocular bioavailability of Bimatoprost for the management of glaucoma.
Collapse
Affiliation(s)
- Sandeep Divate Satyanarayana
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore 575018, India
| | - Amr Selim Abu Lila
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | - Ehssan H Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
- Department of Microbiology and Parasitology, Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum 2404, Sudan
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Hadil Faris Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdul Rahman University, Riyadh 11671, Saudi Arabia
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rompicherla Narayana Charyulu
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore 575018, India
| |
Collapse
|
17
|
Al-Qaysi ZK, Beadham IG, Schwikkard SL, Bear JC, Al-Kinani AA, Alany RG. Sustained release ocular drug delivery systems for glaucoma therapy. Expert Opin Drug Deliv 2023; 20:905-919. [PMID: 37249548 DOI: 10.1080/17425247.2023.2219053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 05/24/2023] [Indexed: 05/31/2023]
Abstract
INTRODUCTION Glaucoma is a group of progressive optic neuropathies resulting in irreversible blindness. It is associated with an elevation of intraocular pressure (>21 mm Hg) and optic nerve damage. Reduction of the intraocular pressure (IOP) through the administration of ocular hypotensive eye drops is one of the most common therapeutic strategies. Patient adherence to conventional eye drops remains a major obstacle in preventing glaucoma progression. Additional problems emerge from inadequate patient education as well as local and systemic side effects associated with adminstering ocular hypotensive drugs. AREAS COVERED Sustained-release drug delivery systems for glaucoma treatment are classified into extraocular systems including wearable ocular surface devices or multi-use (immediate-release) eye formulations (such as aqueous solutions, gels; ocular inserts, contact lenses, periocular rings, or punctual plugs) and intraocular drug delivery systems (such as intraocular implants, and microspheres for supraciliary drug delivery). EXPERT OPINION Sustained release platforms for the delivery of ocular hypotensive drugs (small molecules and biologics) may improve patient adherence and prevent vision loss. Such innovations will only be widely adopted when efficacy and safety has been established through large-scale trials. Sustained release drug delivery can improve glaucoma treatment adherence and reverse/prevent vision deterioration. It is expected that these approaches will improve clinical management and prognosis of glaucoma.
Collapse
Affiliation(s)
- Zinah K Al-Qaysi
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
| | - Ian G Beadham
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
| | - Sianne L Schwikkard
- Department of Chemical and Pharmaceutical Sciences, Kingston University, Kingston Upon Thames, UK
| | - Joseph C Bear
- Department of Chemical and Pharmaceutical Sciences, Kingston University, Kingston Upon Thames, UK
| | - Ali A Al-Kinani
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
| | - Raid G Alany
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
- School of Pharmacy, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
Mostafa M, Al Fatease A, Alany RG, Abdelkader H. Recent Advances of Ocular Drug Delivery Systems: Prominence of Ocular Implants for Chronic Eye Diseases. Pharmaceutics 2023; 15:1746. [PMID: 37376194 PMCID: PMC10302848 DOI: 10.3390/pharmaceutics15061746] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic ocular diseases can seriously impact the eyes and could potentially result in blindness or serious vision loss. According to the most recent data from the WHO, there are more than 2 billion visually impaired people in the world. Therefore, it is pivotal to develop more sophisticated, long-acting drug delivery systems/devices to treat chronic eye conditions. This review covers several drug delivery nanocarriers that can control chronic eye disorders non-invasively. However, most of the developed nanocarriers are still in preclinical or clinical stages. Long-acting drug delivery systems, such as inserts and implants, constitute the majority of the clinically used methods for the treatment of chronic eye diseases due to their steady state release, persistent therapeutic activity, and ability to bypass most ocular barriers. However, implants are considered invasive drug delivery technologies, especially those that are nonbiodegradable. Furthermore, in vitro characterization approaches, although useful, are limited in mimicking or truly representing the in vivo environment. This review focuses on long-acting drug delivery systems (LADDS), particularly implantable drug delivery systems (IDDS), their formulation, methods of characterization, and clinical application for the treatment of eye diseases.
Collapse
Affiliation(s)
- Mahmoud Mostafa
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minya 61519, Egypt;
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62223, Saudi Arabia;
| | - Raid G. Alany
- School of Pharmacy, Kingston University London, Kingston Upon Tames KT1 2EE, UK;
- School of Pharmacy, The University of Auckland, Auckland 1010, New Zealand
| | - Hamdy Abdelkader
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62223, Saudi Arabia;
| |
Collapse
|
19
|
Shen Y, Sun J, Sun X. Intraocular nano-microscale drug delivery systems for glaucoma treatment: design strategies and recent progress. J Nanobiotechnology 2023; 21:84. [PMID: 36899348 PMCID: PMC9999627 DOI: 10.1186/s12951-023-01838-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Glaucoma is a leading cause of irreversible visual impairment and blindness, affecting over 76.0 million people worldwide in 2020, with a predicted increase to 111.8 million by 2040. Hypotensive eye drops remain the gold standard for glaucoma treatment, while inadequate patient adherence to medication regimens and poor bioavailability of drugs to target tissues are major obstacles to effective treatment outcomes. Nano/micro-pharmaceuticals, with diverse spectra and abilities, may represent a hope of removing these obstacles. This review describes a set of intraocular nano/micro drug delivery systems involved in glaucoma treatment. Particularly, it investigates the structures, properties, and preclinical evidence supporting the use of these systems in glaucoma, followed by discussing the route of administration, the design of systems, and factors affecting in vivo performance. Finally, it concludes by highlighting the emerging notion as an attractive approach to address the unmet needs for managing glaucoma.
Collapse
Affiliation(s)
- Yuening Shen
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Jianguo Sun
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, 200031, China.,NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, 200031, China. .,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China.
| |
Collapse
|
20
|
Wu KY, Ashkar S, Jain S, Marchand M, Tran SD. Breaking Barriers in Eye Treatment: Polymeric Nano-Based Drug-Delivery System for Anterior Segment Diseases and Glaucoma. Polymers (Basel) 2023; 15:polym15061373. [PMID: 36987154 PMCID: PMC10054733 DOI: 10.3390/polym15061373] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
The eye has anatomical structures that function as robust static and dynamic barriers, limiting the penetration, residence time, and bioavailability of medications administered topically. The development of polymeric nano-based drug-delivery systems (DDS) could be the solution to these challenges: it can pass through ocular barriers, offering higher bioavailability of administered drugs to targeted tissues that are otherwise inaccessible; it can stay in ocular tissues for longer periods of time, requiring fewer drug administrations; and it can be made up of polymers that are biodegradable and nano-sized, minimizing the undesirable effects of the administered molecules. Therefore, therapeutic innovations in polymeric nano-based DDS have been widely explored for ophthalmic drug-delivery applications. In this review, we will give a comprehensive overview of polymeric nano-based drug-delivery systems (DDS) used in the treatment of ocular diseases. We will then examine the current therapeutic challenges of various ocular diseases and analyze how different types of biopolymers can potentially enhance our therapeutic options. A literature review of the preclinical and clinical studies published between 2017 and 2022 was conducted. Thanks to the advances in polymer science, the ocular DDS has rapidly evolved, showing great promise to help clinicians better manage patients.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Said Ashkar
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Shrieda Jain
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
| | - Michael Marchand
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
- Correspondence:
| |
Collapse
|
21
|
Hashikawa Y, Kato Y, Kaji H, Abe T, Nagai N. A comparative study between a transscleral sustained-release device and eyedrops on intraocular distribution of carteolol hydrochloride. Heliyon 2023; 9:e14392. [PMID: 36942217 PMCID: PMC10023953 DOI: 10.1016/j.heliyon.2023.e14392] [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: 06/27/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
The objectives of this study were to develop a sustained-release device for carteolol hydrochloride (CH) and investigate any potential difference in the intraocular distribution of this agent between the transscleral administration of the device and treatment with eyedrops. The device was formulated with photocurable resin, poly (ethyleneglycol) dimethacrylate, to fit within the curve of the rabbit eyeball. In vitro study showed that CH was released in a sustained-release manner for 2 weeks. The concentration of CH in the retina, choroid/retinal pigment epithelium, sclera, iris, and aqueous humor was determined by high-performance liquid chromatography. Transscleral administration was able to deliver CH to the posterior segment (i.e., retina and choroid/retinal pigment epithelium) rather than the anterior segment (i.e., aqueous humor), while eyedrops delivered CH only to the anterior segment. Transscleral administration could deliver CH to aqueous humor at half the concentration versus treatment with eyedrops and reduced intraocular pressure (IOP) at 1 day after implantation; however, the IOP-lowering effect was not sustained thereafter. In conclusion, transscleral drug delivery may be a useful method for the reduction of IOP. Notably, the aqueous concentration must be equal to that delivered by the eyedrops, and this approach might be preferable for drug delivery to the posterior segment of the eye.
Collapse
Affiliation(s)
- Yoshiko Hashikawa
- Division of Clinical Cell Therapy, United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yuki Kato
- Division of Clinical Cell Therapy, United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hirokazu Kaji
- Department of Biomechanics, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Toshiaki Abe
- Division of Clinical Cell Therapy, United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Nobuhiro Nagai
- Division of Clinical Cell Therapy, United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Corresponding author.
| |
Collapse
|
22
|
Stamer WD, Perkumas KM, Kang MH, Dibas M, Robinson MR, Rhee DJ. Proposed Mechanism of Long-Term Intraocular Pressure Lowering With the Bimatoprost Implant. Invest Ophthalmol Vis Sci 2023; 64:15. [PMID: 36877514 PMCID: PMC10007903 DOI: 10.1167/iovs.64.3.15] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Purpose The purpose of this study was to evaluate the effects of pharmacologically relevant bimatoprost and bimatoprost free acid (BFA) concentrations on matrix metalloproteinase (MMP) gene expression in cells from human aqueous outflow tissues. Methods MMP gene expression by human trabecular meshwork (TM), scleral fibroblast (SF), and ciliary muscle (CM) cells exposed to 10 to 1000 µM bimatoprost or 0.1 to 10 µM BFA (intraocular concentrations after intracameral bimatoprost implant and topical bimatoprost dosing, respectively) was measured by polymerase chain reaction array. Results Bimatoprost dose-dependently upregulated MMP1 and MMP14 mRNA in all cell types and MMP10 and MMP11 mRNA in TM and CM cells; in TM cells from normal eyes, mean MMP1 mRNA levels were 62.9-fold control levels at 1000 µM bimatoprost. BFA upregulated MMP1 mRNA only in TM and SF cells, to two- to three-fold control levels. The largest changes in extracellular matrix (ECM)-related gene expression by TM cells derived from normal (n = 6) or primary open-angle glaucoma (n = 3) eyes occurred with 1000 µM bimatoprost (statistically significant, ≥50% change for 9-11 of 84 genes on the array, versus 1 gene with 10 µM BFA). Conclusions Bimatoprost and BFA had differential effects on MMP/ECM gene expression. Dramatic upregulation in MMP1 and downregulation of fibronectin, which occurred only with bimatoprost at high concentrations observed in bimatoprost implant-treated eyes, may promote sustained outflow tissue remodeling and long-term intraocular pressure reduction beyond the duration of intraocular drug bioavailability. Variability in bimatoprost-stimulated MMP upregulation among cell strains from different donors may help explain differential long-term responses of patients to bimatoprost implant.
Collapse
Affiliation(s)
- W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Kristin M Perkumas
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Min H Kang
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University, Cleveland, Ohio, United States
| | - Mohammed Dibas
- Allergan, an AbbVie company, Irvine, California, United States
| | | | - Douglas J Rhee
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
23
|
Weinreb RN, Bacharach J, Brubaker JW, Medeiros FA, Bejanian M, Bernstein P, Robinson MR. Bimatoprost Implant Biodegradation in the Phase 3, Randomized, 20-Month ARTEMIS Studies. J Ocul Pharmacol Ther 2023; 39:55-62. [PMID: 36378864 PMCID: PMC9885540 DOI: 10.1089/jop.2022.0137] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Purpose: To evaluate the time course of biodegradation of an intracameral, biodegradable, sustained-release bimatoprost implant that lowers intraocular pressure without the need for daily eye drops. Methods: In 2 identically designed, randomized, phase 3 clinical trials, adults with open-angle glaucoma or ocular hypertension and open iridocorneal angles inferiorly in the study eye were administered 10- or 15-μg bimatoprost implant (day 1 and weeks 16 and 32) or twice-daily topical timolol 0.5%. Implants were assessed on gonioscopy throughout the studies. Investigators reported whether implants were visible, estimated the size of visible implants relative to their initial size at implantation, and reported the implant location. Data for 10-μg implant placed on day 1 were pooled from both studies for analysis. Results: A total of 372 patients received the 10-μg bimatoprost implant. The degree of implant biodegradation at each follow-up time point was variable among patients. The implant frequently swelled during the initial phase of biodegradation from 6 to 28 weeks. Accelerated biodegradation occurred between 31 and 52 weeks, resulting in 82% of implants absent or ≤25% of initial size by 52 weeks. By month 20, 95% of implants had biodegraded to absent or ≤25% of initial size. The implant was predominantly located inferiorly in the iridocorneal angle. Conclusions: Bimatoprost implant biodegradation in phase 3 studies showed some degree of variability among patients. Clinically significant implant biodegradation was observed in the majority of patients by 12 months. Clinical studies are in progress to further understand implant biodegradation and the ideal timing for implant re-administration. ClinicalTrials.gov NCT02247804; ClinicalTrials.gov NCT02250651.
Collapse
Affiliation(s)
- Robert N. Weinreb
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, California, USA
| | | | | | | | | | - Paula Bernstein
- Allergan, an AbbVie company, Irvine, California, USA.,Bernstein Biostatistics Consulting, LLC, Laguna Niguel, California, USA
| | - Michael R. Robinson
- Allergan, an AbbVie company, Irvine, California, USA.,Address correspondence to: Dr. Michael R. Robinson, AbbVie, 2525 Dupont Drive, Irvine, CA 92612, USA
| |
Collapse
|
24
|
Asendrych-Wicik K, Zarczuk J, Walaszek K, Ciach T, Markowicz-Piasecka M. Trends in development and quality assessment of pharmaceutical formulations - F2α analogues in the glaucoma treatment. Eur J Pharm Sci 2023; 180:106315. [PMID: 36367507 DOI: 10.1016/j.ejps.2022.106315] [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: 07/04/2022] [Revised: 10/10/2022] [Accepted: 10/27/2022] [Indexed: 12/15/2022]
Abstract
The ocular delivery route presents a number of challenges in terms of drug administration and bioavailability. The low bioavailability following topical ophthalmic administration shows that there is a clear need for in-depth research aimed at finding both more efficacious molecules and formulations precisely targeted at the site of action. Continuous technological development will eventually result in improved bioavailability, lower dosages, reduced toxicity, fewer adverse effects, and thus better patient compliance and treatment efficacy. Technological development, as well as increasingly stringent quality requirements, help stimulate analytical progress. This is also clearly evident in the case of medicinal products used in the treatment of glaucoma, which are the subject of this review. Impurity profiling of PGF2α analogues, either in the pure substance or in the finished formulation, is a crucial step in assessing their quality. The development of specific, accurate and precise stability-indicating analytical methods for determining the content and related substances seems to be an important issue in relation to this tasks. A total of 27 official and in-house analytical methods are presented that are used for the analysis of latanoprost, travoprost and bimatoprost. The conditions for chromatographic separation with UV or MS/MS detection and the available results obtained during method validation are described. In addition, several aspects are discussed, with particular emphasis on the instability of the analogues in aqueous solution and the phenomenon of isomerism, which affects a potentially large number of degradation products.
Collapse
Affiliation(s)
- Katarzyna Asendrych-Wicik
- Analytical Laboratory, Research and Development Department, Polfa Warszawa S.A., ul. Karolkowa 22/24, Warsaw 01-207, Poland; Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Łódź, ul. Muszyńskiego 1, Łódź 90-151, Poland
| | - Jakub Zarczuk
- Liquid Dosage Form Laboratory, Research and Development Department, Polfa Warszawa S.A., Karolkowa 22/24, Warsaw 01-207, Poland; BioMedical Engineering Laboratory Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, ul. Waryńskiego 1, Warsaw 00-645, Poland.
| | - Katarzyna Walaszek
- Technical Research and Development Quality Assurance, Polpharma Bioologics, ul. Spółdzielcza 4, Duchnice 05-850, Poland
| | - Tomasz Ciach
- BioMedical Engineering Laboratory Department of Biotechnology and Bioprocess Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, ul. Waryńskiego 1, Warsaw 00-645, Poland
| | - Magdalena Markowicz-Piasecka
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Łódź, ul. Muszyńskiego 1, Łódź 90-151, Poland
| |
Collapse
|
25
|
Cvenkel B, Kolko M. Devices and Treatments to Address Low Adherence in Glaucoma Patients: A Narrative Review. J Clin Med 2022; 12:jcm12010151. [PMID: 36614952 PMCID: PMC9821329 DOI: 10.3390/jcm12010151] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/05/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Poor adherence to topical glaucoma medications has been linked to worse visual field outcomes in glaucoma patients. Therefore, identifying and overcoming the adherence barriers are expected to slow down the progression of disease. The most common barriers to adherence, in addition to the lack of knowledge, include forgetfulness, side effects of medications, difficulties with drop instillation and low self-efficacy. Symptoms and signs of ocular surface disease, which importantly reduce patients' quality of life, are decreased by using preservative-free topical medications. Sustained drug delivery systems using different vehicles seem promising for relieving the burden of drop administration. Currently, only the bimatoprost sustained-release intracameral implant is available for clinical use and single administration. In the era of digitalization, smart drug delivery-connected devices may aid adherence and, by sharing data with care providers, improve monitoring and adjusting treatment. Selective laser trabeculoplasty as first-line treatment delays the need for drops, whereas minimally invasive glaucoma procedures with and without devices combined with cataract surgery increase the likelihood of patients with early-to-moderate glaucoma to remain drop free or reduce the number of drops needed to control intraocular pressure. The aim of this narrative review is to present and discuss devices and treatments that may improve adherence by reducing the need for drops and side effects of medications and aiding in glaucoma monitoring. For the future, there is a need for studies focusing on clinically important outcomes, quality of life and the cost of intervention with longer post-interventional follow up.
Collapse
Affiliation(s)
- Barbara Cvenkel
- Department of Ophthalmology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
| |
Collapse
|
26
|
Belamkar A, Harris A, Zukerman R, Siesky B, Oddone F, Verticchio Vercellin A, Ciulla TA. Sustained release glaucoma therapies: Novel modalities for overcoming key treatment barriers associated with topical medications. Ann Med 2022; 54:343-358. [PMID: 35076329 PMCID: PMC8794062 DOI: 10.1080/07853890.2021.1955146] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Glaucoma is a progressive optic neuropathy and a leading cause of irreversible blindness. The disease has conventionally been characterized by an elevated intraocular pressure (IOP); however, recent research has built the consensus that glaucoma is not only dependent on IOP but rather represents a multifactorial optic neuropathy. Although many risk factors have been identified ranging from demographics to co-morbidities to ocular structural predispositions, IOP is currently the only modifiable risk factor, most often treated by topical IOP-lowering medications. However, topical hypotensive regimens are prone to non-adherence and are largely inefficient, leading to disease progression in spite of treatment. As a result, several companies are developing sustained release (SR) drug delivery systems as alternatives to topical delivery to potentially overcome these barriers. Currently, Bimatoprost SR (DurystaTM) from Allergan plc is the only FDA-approved SR therapy for POAG. Other SR therapies under investigation include: bimatoprost ocular ring (Allergan) (ClinicalTrials.gov identifier: NCT01915940), iDose® (Glaukos Corporation) (NCT03519386), ENV515 (Envisia Therapeutics) (NCT02371746), OTX-TP (Ocular Therapeutix) (NCT02914509), OTX-TIC (Ocular Therapeutix) (NCT04060144), and latanoprost free acid SR (PolyActiva) (NCT04060758). Additionally, a wide variety of technologies for SR therapeutics are under investigation including ocular surface drug delivery systems such as contact lenses and nanotechnology. While challenges remain for SR drug delivery technology in POAG management, this technology may shift treatment paradigms and dramatically improve outcomes.
Collapse
Affiliation(s)
- Aditya Belamkar
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alon Harris
- Department of Opthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ryan Zukerman
- Department of Opthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Opthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Brent Siesky
- Department of Opthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Thomas A Ciulla
- Indiana University School of Medicine, Indianapolis, IN, USA.,Vitreoretinal Medicine and Surgery, Midwest Eye Institute, Indianapolis, IN, USA
| |
Collapse
|
27
|
Chern KJ, Nettesheim ER, Reid CA, Li NW, Marcoe GJ, Lipinski DM. Prostaglandin-based rAAV-mediated glaucoma gene therapy in Brown Norway rats. Commun Biol 2022; 5:1169. [PMID: 36329259 PMCID: PMC9633612 DOI: 10.1038/s42003-022-04134-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Prostaglandin analogs are first-line treatments for open angle glaucoma and while effective at lowering intraocular pressure, they are undermined by patient non-compliance, causing atrophy of the optic nerve and severe visual impairment. Herein, we evaluate the safety and efficacy of a recombinant adeno-associated viral vector-mediated gene therapy aimed at permanently lowering intraocular pressure through de novo biosynthesis of prostaglandin F2α within the anterior chamber. This study demonstrated a dose dependent reduction in intraocular pressure in normotensive Brown Norway rats maintained over 12-months. Crucially, therapy could be temporarily halted through off-type riboswitch activation, reverting intraocular pressure to normal. Longitudinal multimodal imaging, electrophysiology, and post-mortem histology revealed the therapy was well tolerated at low and medium doses, with no major adverse effects to anterior chamber health, offering a promising alternative to current treatment strategies leading to clinically relevant reductions in intraocular pressure without the need for adherence to a daily treatment regimen.
Collapse
Affiliation(s)
- Kristina J Chern
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Emily R Nettesheim
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher A Reid
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nathan W Li
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Gavin J Marcoe
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Daniel M Lipinski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
28
|
O'Brien Laramy MN, Nagapudi K. Long-acting ocular drug delivery technologies with clinical precedent. Expert Opin Drug Deliv 2022; 19:1285-1301. [PMID: 35912841 DOI: 10.1080/17425247.2022.2108397] [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: 11/04/2022]
Abstract
INTRODUCTION Ocular long-acting injectables and implants (LAIIs) deliver drug at a controlled release rate over weeks to years. A reduced dose frequency eases the treatment burden on patients, minimizes the potential for treatment-related adverse effects, and improves treatment adherence and persistence. AREAS COVERED This review provides a comprehensive landscape of ocular LAII drug delivery technologies with clinical precedent, including eight commercial products and 27 clinical programs. Analysis of this landscape, and the specific technologies with the greatest precedent, provides instructive lessons for researchers interested in this space and insights into the direction of the field. EXPERT OPINION Further technological advancement is required to create biodegradable LAIIs with extended release durations and LAIIs that are compatible with a broader array of therapeutic modalities. In the future, ocular LAII innovations can be applied to diseases with limited treatment options, prophylactic treatment at earlier stages of disease, and cost-effective treatment of ocular diseases in global health settings.
Collapse
Affiliation(s)
- Matthew N O'Brien Laramy
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
29
|
Medeiros FA, Sheybani A, Shah MM, Rivas M, Bai Z, Werts E, Ahmed IIK, Craven ER. Single Administration of Intracameral Bimatoprost Implant 10 µg in Patients with Open-Angle Glaucoma or Ocular Hypertension. Ophthalmol Ther 2022; 11:1517-1537. [PMID: 35643967 PMCID: PMC9253216 DOI: 10.1007/s40123-022-00527-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 12/01/2022] Open
Abstract
Introduction This study evaluated the intraocular pressure (IOP)-lowering efficacy and safety of a single intracameral administration of bimatoprost implant 10 µg in adults with open-angle glaucoma or ocular hypertension. Methods Two identically designed, randomized, 20-month, parallel-group, phase 3 clinical trials (one study eye/patient) compared three administrations of 10- or 15-µg bimatoprost implant (day 1, weeks 16 and 32) with twice-daily topical timolol maleate 0.5%. An open-label, 24-month, phase 1/2 clinical trial compared one or two implants administered in the study eye with once-daily topical bimatoprost 0.03% in the fellow eye. Separate analyses of the pooled phase 3 and phase 1/2 study datasets evaluated outcomes in the 10-µg bimatoprost implant and comparator treatment arms after a single implant administration, up to the time of implant re-administration or rescue with IOP-lowering medication. Results In the phase 3 studies, 10-µg bimatoprost implant single administration demonstrated IOP reductions (hour 0) of 4.9–7.0 mmHg through week 15 from a mean (standard deviation, SD) baseline IOP of 24.5 (2.6) mmHg (n = 374); IOP in the topical timolol BID group was reduced by 6.0–6.3 mmHg from a mean (SD) baseline IOP of 24.5 (2.6) mmHg (n = 373). In the phase 1/2 study (n = 21), median time to use of additional IOP-lowering treatment (Kaplan–Meier analysis) was 273 days (approximately 9 months), and 5 of 21 enrolled patients (23.8%) required no additional IOP-lowering treatment up to 24 months after single administration. In each study, after a single implant administration there were no reports of corneal edema, corneal endothelial cell loss, or corneal touch, and no patients had 20% or greater loss in corneal endothelial cell density. Conclusions Bimatoprost implant single administration lowers IOP and has a favorable safety profile. Additional studies are needed to further evaluate the duration of effect and factors predicting long-term IOP lowering after a single implant administration. Trial registration numbers ClinicalTrials.gov NCT02247804, NCT02250651, and NCT01157364. Supplementary Information The online version contains supplementary material available at 10.1007/s40123-022-00527-6.
Collapse
Affiliation(s)
- Felipe A Medeiros
- Department of Ophthalmology, Duke Eye Center, Duke University, 2351 Erwin Rd, Durham, NC, 27705, USA.
| | - Arsham Sheybani
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | - Erica Werts
- Allergan, an AbbVie Company, Irvine, CA, USA
| | - Iqbal I K Ahmed
- Department of Ophthalmology, University of Toronto, Mississauga, ON, Canada
| | | |
Collapse
|
30
|
Mhatre S, Opere CA, Singh S. Unmet needs in glaucoma therapy: The potential role of hydrogen sulfide and its delivery strategies. J Control Release 2022; 347:256-269. [PMID: 35526614 DOI: 10.1016/j.jconrel.2022.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 12/26/2022]
Abstract
Glaucoma is an optic neuropathy disorder marked by progressive degeneration of the retinal ganglion cells (RGC). It is a leading cause of blindness worldwide, prevailing in around 2.2% of the global population. The hallmark of glaucoma, intraocular pressure (IOP), is governed by the aqueous humor dynamics which plays a crucial role in the pathophysiology of the diesease. Glaucomatous eye has an IOP of more than 22 mmHg as compared to normotensive pressure of 10-21 mmHg. Currently used treatments focus on reducing the elevated IOP through use of classes of drugs that either increase aqueous humor outflow and/or decrease its production. However, effective treatments should not only reduce IOP, but also offer neuroprotection and regeneration of RGCs. Hydrogen Sulfide (H2S), a gasotransmitter with several endogenous functions in mammalian tissues, is being investigated for its potential application in glaucoma. In addition to decreasing IOP by increasing aqueous humor outflow, it scavenges reactive oxygen species, upregulates the cellular antioxidant glutathione and protects RGCs from excitotoxicity. Despite the potential of H2S in glaucoma, its delivery to anterior and posterior regions of the eye is a challenge due to its unique physicochemical properties. Firstly, development of any delivery system should not require an aqueous environment since many H2S donors are susceptible to burst release of the gas in contact with water, causing potential toxicity and adverse effects owing to its inherent toxicity at higher concentrations. Secondly, the release of the gas from the donor needs to be sustained for a prolonged period of time to reduce dosing frequency as per the requirements of regulatory bodies. Lastly, the delivery system should provide adequate bioavailability throughout its period of application. Hence, an ideal delivery system should aim to tackle all the above challenges related to barriers of ocular delivery and physicochemical properties of H2S itself. This review discusses the therapeutic potential of H2S, its delivery challenges and strategies to overcome the associated chalenges.
Collapse
Affiliation(s)
- Susmit Mhatre
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA.
| | - Catherine A Opere
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA.
| | - Somnath Singh
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA.
| |
Collapse
|
31
|
Calpains as mechanistic drivers and therapeutic targets for ocular disease. Trends Mol Med 2022; 28:644-661. [PMID: 35641420 PMCID: PMC9345745 DOI: 10.1016/j.molmed.2022.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022]
Abstract
Ophthalmic neurodegenerative diseases encompass a wide array of molecular pathologies unified by calpain dysregulation. Calpains are calcium-dependent proteases that perpetuate cellular death and inflammation when hyperactivated. Calpain inhibition trials in other organs have faced pharmacological challenges, but the eye offers many advantages for the development and testing of targeted molecular therapeutics, including small molecules, peptides, engineered proteins, drug implants, and gene-based therapies. This review highlights structural mechanisms underlying calpain activation, distinct cellular expression patterns, and in vivo models that link calpain hyperactivity to human retinal and developmental disease. Optimizing therapeutic approaches for calpain-mediated eye diseases can help accelerate clinically feasible strategies for treating calpain dysregulation in other diseased tissues.
Collapse
|
32
|
Ghosn C, Rajagopalan L, Ugarte S, Mistry S, Orilla W, Goodkin ML, Robinson MR, Engles M, Dibas M. Intraocular Pressure-Lowering Efficacy of a Sustained-Release Bimatoprost Implant in Dog Eyes Pretreated with Selective Laser Trabeculoplasty. J Ocul Pharmacol Ther 2022; 38:311-318. [PMID: 35442770 PMCID: PMC9125576 DOI: 10.1089/jop.2021.0104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/04/2022] [Indexed: 02/03/2023] Open
Abstract
Purpose: To assess the intraocular pressure (IOP)-lowering effect of a biodegradable bimatoprost implant following selective laser trabeculoplasty (SLT) in a canine model. Methods: Unilateral SLT was performed in 11 normotensive, treatment-naive beagle dogs. IOP was measured at baseline (pre-SLT) and weekly post-SLT (≤10 weeks). After IOP returned to baseline or at 10 weeks (whichever occurred first), a sustained-release bimatoprost implant was administered bilaterally in the anterior chamber of each animal. IOP was measured weekly for 4 weeks and then every 2 weeks up to week 42. Results: The main outcomes included the IOP change (%) from baseline, calculated in both eyes in the overall population, SLT responder subgroup (defined by peak IOP reduction from baseline ≥3 mmHg or ≥15% for >1 week post-SLT), and SLT nonresponder subgroup (defined by peak IOP reduction from baseline <3 mmHg or <15%). The bimatoprost implant lowered IOP similarly in both the SLT-treated and fellow SLT-naive eyes. Following bimatoprost implant administration, the mean (standard deviation [SD]) peak IOP reduction from baseline was 34.4% (8.5%) in SLT-treated eyes and 35.7% (5.9%) in fellow SLT-naive eyes. The bimatoprost implant lowered IOP comparably (P > 0.17) in eyes that responded to SLT (mean [SD] peak IOP reduction, 34.6% [10.7%]; n = 6) and those that did not (mean [SD] peak IOP reduction, 34.1% [6.1%]; n = 5). Conclusion: The bimatoprost implant effectively lowered IOP in eyes pretreated with SLT, regardless of response to SLT. The current data suggest that eyes previously treated with SLT can still benefit from the intracameral bimatoprost implant.
Collapse
Affiliation(s)
- Corine Ghosn
- Allergan, An AbbVie Company, Irvine, California, USA
| | | | - Sveti Ugarte
- Allergan, An AbbVie Company, Irvine, California, USA
| | - Shruti Mistry
- Allergan, An AbbVie Company, Irvine, California, USA
| | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Bacharach J, Tatham A, Ferguson G, Belalcázar S, Thieme H, Goodkin ML, Chen MY, Guo Q, Liu J, Robinson MR, Bejanian M, Wirta DL. Phase 3, Randomized, 20-Month Study of the Efficacy and Safety of Bimatoprost Implant in Patients with Open-Angle Glaucoma and Ocular Hypertension (ARTEMIS 2). Drugs 2021; 81:2017-2033. [PMID: 34724172 PMCID: PMC8602154 DOI: 10.1007/s40265-021-01624-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2021] [Indexed: 11/24/2022]
Abstract
Objective To evaluate the intraocular pressure (IOP)-lowering efficacy and safety of 10 and 15 µg bimatoprost implant in patients with open-angle glaucoma (OAG) or ocular hypertension (OHT). Methods This randomized, 20-month, multicenter, masked, parallel-group, phase 3 trial enrolled 528 patients with OAG or OHT and an open iridocorneal angle inferiorly in the study eye. Study eyes were administered 10 or 15 µg bimatoprost implant on day 1, week 16, and week 32, or twice-daily topical timolol maleate 0.5%. Primary endpoints were IOP and IOP change from baseline through week 12. Safety measures included treatment-emergent adverse events (TEAEs) and corneal endothelial cell density (CECD). Results Both 10 and 15 µg bimatoprost implant met the primary endpoint of noninferiority to timolol in IOP lowering through 12 weeks. Mean IOP reductions from baseline ranged from 6.2–7.4, 6.5–7.8, and 6.1–6.7 mmHg through week 12 in the 10 µg implant, 15 µg implant, and timolol groups, respectively. IOP lowering was similar after the second and third implant administrations. Probabilities of requiring no IOP-lowering treatment for 1 year after the third administration were 77.5% (10 µg implant) and 79.0% (15 µg implant). The most common TEAE was conjunctival hyperemia, typically temporally associated with the administration procedure. Corneal TEAEs of interest (primarily corneal endothelial cell loss, corneal edema, and corneal touch) were more frequent with the 15 than the 10 µg implant and generally were reported after repeated administrations. Loss in mean CECD from baseline to month 20 was ~ 5% in 10 µg implant-treated eyes and ~ 1% in topical timolol-treated eyes. Visual field progression (change in the mean deviation from baseline) was reduced in the 10 µg implant group compared with the timolol group. Conclusions The results corroborated the previous phase 3 study of the bimatoprost implant. The bimatoprost implant met the primary endpoint and effectively lowered IOP. The majority of patients required no additional treatment for 12 months after the third administration. The benefit-risk assessment favored the 10 over the 15 µg implant. Studies evaluating other administration regimens with reduced risk of corneal events are ongoing. The bimatoprost implant has the potential to improve adherence and reduce treatment burden in glaucoma. Clinicaltrials.gov Identifier NCT02250651. Supplementary Information The online version contains supplementary material available at 10.1007/s40265-021-01624-9.
Collapse
Affiliation(s)
- Jason Bacharach
- North Bay Eye Associates, 104 Lynch Creek Way, Suite 12, Petaluma, CA, 94954, USA.
| | | | | | | | - Hagen Thieme
- University Eye Clinic Magdeburg, Magdeburg, Germany
| | | | | | - Qiang Guo
- Allergan, an AbbVie company, Irvine, CA, USA
| | - Jeen Liu
- Allergan, an AbbVie company, Irvine, CA, USA
| | | | | | - David L Wirta
- Aesthetic Eye Care Institute and Eye Research Foundation, Newport Beach, CA, USA
| |
Collapse
|
35
|
Abstract
Background: Sustained release drug delivery has the potential to change glaucoma care by decreasing the challenge of medication adherence. Many approaches are in development, but this review focuses on Durysta (Allergan plc, Dublin, Ireland), the only FDA-approved sustained release intracameral treatment available at this time. Key Findings: Durysta is a bimatoprost sustained release (BimSR) intracameral implant. Clinical trials have demonstrated that BimSR implants can provide comparable levels of intraocular pressure (IOP) control as topical eyedrops. BimSR has advantages such as decreasing concerns regarding drop adherence, reducing ocular surface and periocular side effects from topical drops, and decreased daily treatment burden for patients. In addition, studies have shown continued IOP lowering in some eyes during extended follow-up periods when all of the BimSR medication has already been delivered. Hypothesized mechanisms to explain this finding include increased matrix metalloproteinase expression that causes extracellular matrix reorganization to permit greater aqueous outflow, as well as decreased episcleral venous pressure. The major safety concern at this time for Durysta and future intracameral implants is corneal endothelial cell loss, which was worse with repeat BimSR administration compared to single dosing. Several studies are underway to investigate mechanisms of action and to better understand safe and effective dosing of medications in this class.
Collapse
Affiliation(s)
| | - Michael M Lin
- Glaucoma Service, Massachusetts Eye and Ear, Boston, MA, USA
| |
Collapse
|
36
|
Soliman K, Jirjees F, Sonawane R, Sheshala R, Wang Y, Jones D, Singh TRR. Latanoprost Quantification in Ocular Implants and Tissues: HPLC-Fluorescence vs HPLC-UV. J Chromatogr Sci 2021; 59:64-70. [PMID: 33047781 DOI: 10.1093/chromsci/bmaa078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 06/20/2020] [Accepted: 09/12/2020] [Indexed: 11/14/2022]
Abstract
Anti-glaucoma latanoprost-loaded ocular implants provide prolonged delivery and enhanced bioavailability relative to the conventional eye drops. This study aims at the development and validation of a reversed-phase high-performance liquid chromatography method for quantitative analysis of nanogram levels of latanoprost in the eye, and for the first time, compares the use of fluorescence vs ultraviolet (UV) detectors in latanoprost quantification. The mobile phase was composed of acetonitrile:0.1% v/v formic acid (60:40, v/v) with a flow rate of 1 mL/min and separation was done using a C18 column at temperature 40°C. The fluorescence excitation and emission wavelengths were set at 265 and 285 nm, respectively, while the UV absorption was measured at 200 nm. The latanoprost concentration-peak area relationship maintained its linearity (R2 = 0.9999) over concentration ranges of 0.063-10 μg/mL and 0.212-10 μg/mL for the fluorescence and UV detectors, respectively. The UV detector showed better precision, while the fluorescence detector exhibited higher robustness and greater sensitivity, with a detection limit of 0.021 μg/mL. The fluorescence detector was selected for quantification of latanoprost released from ocular implants in vitro and in porcine ocular tissues. The developed method is a robust, rapid and cost-effective alternative to liquid chromatography-mass spectrometry for routine analysis of latanoprost released from ocular implants.
Collapse
Affiliation(s)
- Karim Soliman
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.,Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Feras Jirjees
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.,Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.,College of Pharmacy, University of Sharjah, University City Rd, Sharjah, UAE
| | - Rahul Sonawane
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.,Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ravi Sheshala
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300 Puncak Alam, Kuala Selangor, Malaysia
| | - Yujing Wang
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.,Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - David Jones
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.,Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Thakur Raghu Raj Singh
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.,Re-Vana Therapeutics, McClay Research Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| |
Collapse
|
37
|
Kompella UB, Hartman RR, Patil MA. Extraocular, periocular, and intraocular routes for sustained drug delivery for glaucoma. Prog Retin Eye Res 2021; 82:100901. [PMID: 32891866 PMCID: PMC8317199 DOI: 10.1016/j.preteyeres.2020.100901] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 08/22/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
Although once daily anti-glaucoma drug therapy is a current clinical reality, most therapies require multiple dosing and there is an unmet need to develop convenient, safe, and effective sustained release drug delivery systems for long-term treatment to improve patient adherence and outcomes. One of the first sustained release drug delivery systems was approved for the reduction of intraocular pressure in glaucoma patients. It is a polymeric reservoir-type insert delivery system, Ocusert™, placed under the eyelid and on the ocular surface for zero-order drug release over one week. The insert, marketed in two strengths, released pilocarpine on the eye surface. While many clinicians appreciated this drug product, it was eventually discontinued. No similar sustained release non-invasive drug delivery system has made it to the market to date for treating glaucoma. Drug delivery systems under development include punctal plugs, ring-type systems, contact lenses, implants, microspheres, nanospheres, gels, and other depot systems placed in the extraocular, periocular, or intraocular regions including intracameral, supraciliary, and intravitreal spaces. This article discusses the advantages and disadvantages of the various routes of administration and delivery systems for sustained glaucoma therapy. It also provides the reader with some examples and discussion of drug delivery systems that could potentially be applied for glaucoma treatment. Interestingly, one intracamerally injected implant, Durysta™, was approved recently for sustained intraocular pressure reduction. However, long-term acceptance of such devices has yet to be established. The ultimate success of the delivery system will depend on efficacy relative to eye drop dosing, safety, reimbursement options, and patient acceptance. Cautious development efforts are warranted considering prior failed approaches for sustained glaucoma drug delivery. Neuroprotective approaches for glaucoma therapy including cell, gene, protein, and drug-combination therapies, mostly administered intravitreally, are also rapidly progressing towards assessment in humans.
Collapse
Affiliation(s)
- Uday B Kompella
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Rachel R Hartman
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Madhoosudan A Patil
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
38
|
Macoon R, Chauhan A. Ophthalmic delivery of hydrophilic drugs through drug-loaded oleogels. Eur J Pharm Sci 2021; 158:105634. [DOI: 10.1016/j.ejps.2020.105634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 10/23/2022]
|
39
|
New strategies for the management of ocular surface disease in glaucoma patients. Curr Opin Ophthalmol 2021; 32:134-140. [PMID: 33492867 DOI: 10.1097/icu.0000000000000739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Glaucoma patients commonly suffer from ocular surface disease (OSD). As treatment strategies, medications and devices for the treatment of OSD as well as glaucoma surgical approaches evolve rapidly, it is important to consider their application to these patients. RECENT FINDINGS OSD in glaucoma patients may lead to reduced reliability of diagnostic tests, decreased medication compliance, poor surgical outcomes, and overall decreased quality of life. Chronic use of topical glaucoma medications has been linked to the development of limbal stem cell deficiency, and the role of preservatives in OSD continues to be demonstrated. Preservative free glaucoma medications as well as new anti-inflammatory agents for the treatment of OSD are now available. Omega-3 fatty acid supplementation and punctal plugs have been shown to benefit glaucoma patients with OSD. Drop burden may be reduced through the use of the new sustained-release delivery systems, selective laser trabeculoplasty, and minimally invasive glaucoma surgery. SUMMARY There are multiple emerging strategies for managing OSD that may be applied to patients with glaucoma. With continued research and clinical experiences, we hope to better understand the multifaceted relationship between glaucoma and OSD and develop evidence-based algorithms for the management of these complex patients.
Collapse
|
40
|
Smith SM, Salmon JH, Abbaraju S, Amin R, Gilger BC. Tolerability, pharmacokinetics, and pharmacodynamics of a brinzolamide episcleral sustained release implant in normotensive New Zealand white rabbits. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
41
|
Kesav NP, Young CEC, Ertel MK, Seibold LK, Kahook MY. Sustained-release drug delivery systems for the treatment of glaucoma. Int J Ophthalmol 2021; 14:148-159. [PMID: 33469497 PMCID: PMC7790669 DOI: 10.18240/ijo.2021.01.21] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022] Open
Abstract
Glaucoma, a leading cause of irreversible blindness, affects more than 64 million people worldwide and is expected to grow in number due to the aging global population and enhanced methods of detection. Although topical therapies are often effective when used as prescribed, the drawbacks of current medical management methods include poor patient adherence, local and systemic side effects, and in some cases, limited therapeutic efficacy. Novel ocular drug delivery platforms promise to deliver differentiated drug formulations with targeted delivery leveraging patient-independent administration. Several platforms are in various stages of development with promising pre-clinical and clinical data. The Bimatoprost Sustained Release (SR) intracameral implant was approved in the United States in March of 2020, making it the first long-term injectable therapy available for the treatment of glaucoma. This review aims to provide an update on novel sustained release drug delivery systems that are available today as well as those that might be commercialized in coming years.
Collapse
Affiliation(s)
- Natasha P. Kesav
- Department of Ophthalmology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | | | - Monica K. Ertel
- Department of Ophthalmology, University of Colorado School of Medicine, CO 80045, USA
| | - Leonard K. Seibold
- Department of Ophthalmology, University of Colorado School of Medicine, CO 80045, USA
| | - Malik Y. Kahook
- Department of Ophthalmology, University of Colorado School of Medicine, CO 80045, USA
| |
Collapse
|
42
|
Abstract
Abstract
Background
Polymers are essential components of many drug delivery systems and biomedical products. Despite the utility of many currently available polymers, there exists a demand for materials with improved characteristics and functionality. Due to the extensive safety testing required for new excipient approval, the introduction and use of new polymers is considerably limited. The blending of currently approved polymers provides a valuable solution by which the limitations of individual polymers can be addressed.
Main body
Polymer blends combine two or more polymers resulting in improved, augmented, or customized properties and functionality which can result in significant advantages in drug delivery applications. This review discusses the rationale for the use of polymer blends and blend polymer-polymer interactions. It provides examples of their use in commercially marketed products and drug delivery systems. Examples of polymer blends in amorphous solid dispersions and biodegradable systems are also discussed. A classification scheme for polymer blends based on the level of material processing and interaction is presented.
Conclusion
The use of polymer blends represents a valuable and under-utilized resource in addressing a diverse range of drug delivery challenges. It is anticipated that new drug molecule development challenges such as bioavailability enhancement and the demand for enabling excipients will lead to increased applications of polymer blends in pharmaceutical products.
Graphical abstract
Collapse
|
43
|
Miller PE, Eaton JS. Medical anti-glaucoma therapy: Beyond the drop. Vet Ophthalmol 2020; 24 Suppl 1:2-15. [PMID: 33164328 DOI: 10.1111/vop.12843] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/30/2020] [Accepted: 10/20/2020] [Indexed: 12/25/2022]
Abstract
Barriers to effective medical therapy are numerous and include difficulties with effective and sustained control of intraocular pressure (IOP) and adherence to prescribed anti-glaucoma drop regimens. In an effort to circumvent these challenges, a number of new anti-glaucoma therapies with sustained effects have emerged. Methods for sustained delivery of prostaglandin analogs are being intensely investigated and many are in human clinical trials. Intracameral devices include the following: Allergan's Durysta™ Bimatoprost SR, Envisia Therapeutics' ENV515 travoprost implant, Glaukos' iDose™ , Ocular Therapeutix's OTX-TIC travoprost implant, and Santen's polycaprolactone implant with PGE2-derivative DE-117. Other prostaglandin-based technologies include Allergan's bimatoprost ring (placed in the conjunctival fornix), Ocular Therapeutics' OTX-TP intracanalicular travoprost implant, subconjunctival latanoprost in a liposomal formulation, and the PGE2 derivative PGN 9856-isopropyl ester that is applied to the periorbital skin. Exciting breakthroughs in gene therapy include using viral vectors to correct defective genes such as MYOC or to modulate gonioimplant fibrosis, CRISPR technology to edit MYOC or to alter aquaporin to reduce aqueous humor production, and siRNA technology to silence specific genes. Stem cell technology can repopulate depleted tissues or, in the case of Neurotech's Renexus® NT-501 intravitreal implant, serve as a living drug delivery device that continuously secretes neurotrophic factors. Other unique approaches involve nanotechnology, nasal sprays that deliver drug directly to the optic nerve and noninvasive alternating current stimulation of surviving cells in the optic nerve. Over time these modalities are likely to challenge the preeminent role that drops currently play in the medical treatment of glaucoma in animals.
Collapse
Affiliation(s)
- Paul E Miller
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua Seth Eaton
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
44
|
Craven ER, Walters T, Christie WC, Day DG, Lewis RA, Goodkin ML, Chen M, Wangsadipura V, Robinson MR, Bejanian M. 24-Month Phase I/II Clinical Trial of Bimatoprost Sustained-Release Implant (Bimatoprost SR) in Glaucoma Patients. Drugs 2020; 80:167-179. [PMID: 31884564 PMCID: PMC7007425 DOI: 10.1007/s40265-019-01248-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective The objective of this study was to evaluate the safety and intraocular pressure (IOP)-lowering effects over 24 months of biodegradable bimatoprost sustained-release implant (Bimatoprost SR) administration versus topical bimatoprost 0.03% in patients with open-angle glaucoma (OAG). Methods This was a phase I/II, prospective, 24-month, dose-ranging, paired-eye controlled clinical trial. At baseline following washout, adult patients with OAG (N = 75) received Bimatoprost SR (6, 10, 15, or 20 µg) intracamerally in the study eye; the fellow eye received topical bimatoprost 0.03% once daily. Rescue topical IOP-lowering medication or single repeat administration with implant was permitted. The primary endpoint was IOP change from baseline. Safety measures included adverse events (AEs). Results At month 24, mean IOP reduction from baseline was 7.5, 7.3, 7.3, and 8.9 mmHg in eyes treated with Bimatoprost SR 6, 10, 15, and 20 µg, respectively, versus 8.2 mmHg in pooled fellow eyes; 68, 40, and 28% of pooled study eyes had not been rescued/retreated at months 6, 12, and 24, respectively. AEs in study eyes that occurred ≤ 2 days post-procedure typically were transient. After 2 days post-procedure, overall AE incidence was similar between study and fellow eyes, with some events typically associated with topical prostaglandin analogs having lower incidence in study eyes. Conclusions Bimatoprost SR showed favorable efficacy and safety profiles up to 24 months, with all evaluated dose strengths demonstrating overall IOP-reducing effects comparable to those of topical bimatoprost. Targeted and sustained delivery of bimatoprost resulted in protracted IOP lowering, suggesting that Bimatoprost SR may represent a transformational new approach to glaucoma therapy. Clinicaltrials.gov identifier: NCT01157364 Electronic supplementary material The online version of this article (10.1007/s40265-019-01248-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- E Randy Craven
- Johns Hopkins University School of Medicine, 600 N. Wolfe Street, 110, Baltimore, MD, 21287, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Short B. Selected Aspects of Ocular Toxicity Studies With a Focus on High-Quality Pathology Reports: A Pathology/Toxicology Consultant's Perspective. Toxicol Pathol 2020; 49:673-699. [PMID: 32815474 DOI: 10.1177/0192623320946712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ocular toxicity studies are the bedrock of nonclinical ocular drug and drug-device development, and there has been an evolution in experience, technologies, and challenges to address that ensures safe clinical trials and marketing authorization. The expectations of a well-designed ocular toxicity study and the generation of a coherent, integrative ocular toxicology report and subreports are high, and this article provides a pathology/toxicology consultant's perspective on achieving that goal. The first objective is to cover selected aspects of study designs for ocular toxicity studies including considerations for contract research organization selection, minipig species selection, unilateral versus bilateral dosing, and in-life parameters based on fit-for-purpose study objectives. The main objective is a focus on a high-quality ocular pathology report that includes ocular histology procedures to meet regulatory expectations and a report narrative and tables that correlate microscopic findings with key ophthalmic findings and presents a clear interpretation of test article-, vehicle-, and procedure-related ocular and extraocular findings with identification of adversity and a pathology peer review. The last objective covers considerations for a high-quality ophthalmology report, which in concert with a high-quality pathology report, will pave the way for a best quality toxicology report for an ocular toxicity study.
Collapse
Affiliation(s)
- Brian Short
- Brian Short Consulting, LLC, Laguna Beach, CA, USA
| |
Collapse
|
46
|
Shen J, Moats RA, Pollack HA, Robinson MR, Attar M. Distribution of 14C-Latanoprost Following a Single Intracameral Administration Versus Repeated Topical Administration. Ophthalmol Ther 2020; 9:929-940. [PMID: 32740740 PMCID: PMC7708611 DOI: 10.1007/s40123-020-00285-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Indexed: 11/29/2022] Open
Abstract
Purpose To qualitatively evaluate the ocular and periocular distribution of 14C-latanoprost following a single intracameral administration or repeated topical ocular administration in beagle dogs and cynomolgus monkeys. Methods In the dog study, three animals received an intracameral dose of 14C-latanoprost bilaterally and were euthanized at 1, 2, and 4 h post dose; three control animals received topical 14C-latanoprost bilaterally once daily for 5 days and were euthanized at 1, 4, and 24 h post final dose. Sagittal 40-µm sections of eyes with surrounding tissues were collected and processed for autoradiography. Methods in the monkey study were similar; two animals received a unilateral intracameral dose of 14C-latanoprost. Results After intracameral dosing in dogs, radioactivity was concentrated in the cornea, iris, ciliary body, and anterior chamber with no radioactivity detected in the eyelids or other periorbital tissues. After topical dosing, radioactivity was distributed in the bulbar conjunctiva, cornea, anterior chamber, iris, ciliary body, upper and lower eyelids, and periorbital tissues (fat/muscle). After intracameral dosing in monkeys, radioactivity was concentrated in the anterior chamber, cornea, iris, ciliary body, and posteriorly along the uveoscleral outflow pathway; there was no radioactivity in the eyelids or periorbital tissues aside from signal in the nasolacrimal duct, likely from reflux of 14C-latanoprost into the tear film. Conclusions Intracameral delivery resulted in more selective target tissue drug exposure. Intracameral drug delivery has potential to reduce ocular surface and periocular adverse effects associated with topical administration of prostaglandin analogues, such as eyelash growth and periorbital fat atrophy. Electronic supplementary material The online version of this article (10.1007/s40123-020-00285-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Shen
- Allergan, An AbbVie Company, Irvine, CA, USA.
| | - Rex A Moats
- Moats Laboratory at the Saban Research Institute, Children's Hospital Los Angeles Research Imaging Core, Department of Radiology, Keck School of Medicine USC, Los Angeles, CA, USA
| | - Harvey A Pollack
- Moats Laboratory at the Saban Research Institute, Children's Hospital Los Angeles Research Imaging Core, Department of Radiology, Keck School of Medicine USC, Los Angeles, CA, USA
| | | | | |
Collapse
|
47
|
Mietzner R, Kade C, Froemel F, Pauly D, Stamer WD, Ohlmann A, Wegener J, Fuchshofer R, Breunig M. Fasudil Loaded PLGA Microspheres as Potential Intravitreal Depot Formulation for Glaucoma Therapy. Pharmaceutics 2020; 12:pharmaceutics12080706. [PMID: 32727014 PMCID: PMC7464914 DOI: 10.3390/pharmaceutics12080706] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Rho-associated protein kinase (ROCK) inhibitors allow for causative glaucoma therapy. Unfortunately, topically applied ROCK inhibitors suffer from high incidence of hyperemia and low intraocular bioavailability. Therefore, we propose the use of poly (lactide-co-glycolide) (PLGA) microspheres as a depot formulation for intravitreal injection to supply outflow tissues with the ROCK inhibitor fasudil over a prolonged time. Fasudil-loaded microspheres were prepared by double emulsion solvent evaporation technique. The chemical integrity of released fasudil was confirmed by mass spectrometry. The biological activity was measured in cell-based assays using trabecular meshwork cells (TM cells), Schlemm’s canal cells (SC cells), fibroblasts and adult retinal pigment epithelium cells (ARPE-19). Cellular response to fasudil after its diffusion through vitreous humor was investigated by electric cell-substrate impedance sensing. Microspheres ranged in size from 3 to 67 µm. The release of fasudil from microspheres was controllable and sustained for up to 45 days. Released fasudil reduced actin stress fibers in TM cells, SC cells and fibroblasts. Decreased collagen gel contraction provoked by fasudil was detected in TM cells (~2.4-fold), SC cells (~1.4-fold) and fibroblasts (~1.3-fold). In addition, fasudil readily diffused through vitreous humor reaching its target compartment and eliciting effects on TM cells. No negative effects on ARPE-19 cells were observed. Since fasudil readily diffuses through the vitreous humor, we suggest that an intravitreal drug depot of ROCK inhibitors could significantly improve current glaucoma therapy particularly for patients with comorbid retinal diseases.
Collapse
Affiliation(s)
- Raphael Mietzner
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany;
| | - Christian Kade
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; (C.K.); (J.W.)
| | - Franziska Froemel
- Department of Human Anatomy and Embryology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; (F.F.); (R.F.)
| | - Diana Pauly
- Experimental Ophthalmology, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany;
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA;
| | - Andreas Ohlmann
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, Mathildenstrasse 8, 80336 Munich, Germany;
| | - Joachim Wegener
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; (C.K.); (J.W.)
- Fraunhofer Research Institution for Microsystems and Solid State Technologies EMFT, Universitaetsstrasse 31, 93040 Regensburg, Germany
| | - Rudolf Fuchshofer
- Department of Human Anatomy and Embryology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany; (F.F.); (R.F.)
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93040 Regensburg, Germany;
- Correspondence: ; Tel.: +49-(0)-941-943-4828
| |
Collapse
|
48
|
Lee SS, Robinson MR, Weinreb RN. Episcleral Venous Pressure and the Ocular Hypotensive Effects of Topical and Intracameral Prostaglandin Analogs. J Glaucoma 2020; 28:846-857. [PMID: 31261285 PMCID: PMC6735525 DOI: 10.1097/ijg.0000000000001307] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
There is a limit beyond which increasing either the concentration of a prostaglandin analog (PGA) or its dosing frequency fails to produce increases in ocular hypotensive efficacy with topical dosing. Intracameral PGA dosing with a bimatoprost implant, however, does not exhibit the same intraocular pressure (IOP)-lowering plateau at studied concentrations, and the maximum-achievable ocular hypotensive effects are not yet known. This suggests that the bimatoprost intracameral implant may activate another mechanism of action in addition to the mechanism(s) activated by topical application. Episcleral venous pressure (EVP) is a key determinant of IOP, and experimental manipulation of the episcleral vasculature can change both EVP and IOP. The recent observation that topical and intracameral PGA drug delivery routes produce different patterns of conjunctival hyperemia suggested that the differences in the IOP-lowering profiles may be caused by differing effects on the episcleral vasculature. Recent experiments in animals have shown that topical PGAs increase EVP, while the bimatoprost intracameral implant causes a smaller, transient increase in EVP, followed by a sustained decrease. The increase in EVP could be limiting the IOP-lowering efficacy of topical PGAs. In contrast, the decrease in EVP associated with the bimatoprost implant could explain its enhanced IOP-lowering effects. Further research on EVP as a target for IOP lowering is indicated to improve our understanding of this potentially important pathway for treating patients with glaucoma.
Collapse
Affiliation(s)
| | | | - Robert N Weinreb
- Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, La Jolla, CA
| |
Collapse
|
49
|
Industry update: the latest developments in the field of therapeutic delivery, March 2020. Ther Deliv 2020; 11:471-484. [PMID: 32594855 DOI: 10.4155/tde-2020-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
50
|
Medeiros FA, Walters TR, Kolko M, Coote M, Bejanian M, Goodkin ML, Guo Q, Zhang J, Robinson MR, Weinreb RN. Phase 3, Randomized, 20-Month Study of Bimatoprost Implant in Open-Angle Glaucoma and Ocular Hypertension (ARTEMIS 1). Ophthalmology 2020; 127:1627-1641. [PMID: 32544560 DOI: 10.1016/j.ophtha.2020.06.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 10/24/2022] Open
Abstract
PURPOSE To evaluate the intraocular pressure (IOP)-lowering efficacy and safety of 10- and 15-μg bimatoprost implant in subjects with open-angle glaucoma (OAG) and ocular hypertension (OHT) after initial and repeated administrations. DESIGN Randomized, 20-month, multicenter, subject- and efficacy evaluator-masked, parallel-group, phase 3 clinical study. PARTICIPANTS Adults with OAG or OHT in each eye, open iridocorneal angle inferiorly in the study eye, and study eye baseline IOP (hour 0; 8 am) of 22-32 mmHg after washout. METHODS Study eyes received bimatoprost implant 10 μg (n = 198) or 15 μg (n = 198) on day 1 with readministration at weeks 16 and 32, or twice-daily topical timolol maleate 0.5% (n = 198). Intraocular pressure was measured at hours 0 and 2 at each visit. MAIN OUTCOME MEASURES Primary end points were IOP and change from baseline IOP through week 12. Safety measures included treatment-emergent adverse events (TEAEs) and corneal endothelial cell density (CECD). RESULTS Both dose strengths of bimatoprost implant were noninferior to timolol in IOP lowering after each administration. Mean diurnal IOP was 24.0, 24.2, and 23.9 mmHg at baseline and from 16.5-17.2, 16.5-17.0, and 17.1-17.5 mmHg through week 12 in the 10-μg implant, 15-μg implant, and timolol groups, respectively. The incidence of corneal and inflammatory TEAEs of interest (e.g., corneal endothelial cell loss, iritis) was higher with bimatoprost implant than timolol and highest with the 15-μg dose strength. Incidence of corneal TEAEs increased after repeated treatment; with 3 administrations at fixed 16-week intervals, incidence of ≥20% CECD loss was 10.2% (10-μg implant) and 21.8% (15-μg implant). Mean best-corrected visual acuity (BCVA) was stable; 3 implant-treated subjects with corneal TEAEs had >2-line BCVA loss at their last visit. CONCLUSIONS Both dose strengths of bimatoprost implant met the primary end point of noninferiority to timolol through week 12. One year after 3 administrations, IOP was controlled in most subjects without additional treatment. The risk-benefit assessment favored the 10-μg implant over the 15-μg implant. Ongoing studies are evaluating other administration regimens to reduce the potential for CECD loss. The bimatoprost implant has potential to improve adherence and reduce treatment burden in glaucoma.
Collapse
Affiliation(s)
- Felipe A Medeiros
- Department of Ophthalmology, Duke University, Durham, North Carolina
| | | | - Miriam Kolko
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Coote
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | | | | | | | | | | | - Robert N Weinreb
- Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, California
| | | |
Collapse
|