1
|
Wang W, Wang H. Understanding the complex genetics and molecular mechanisms underlying glaucoma. Mol Aspects Med 2023; 94:101220. [PMID: 37856931 DOI: 10.1016/j.mam.2023.101220] [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: 06/29/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/21/2023]
Abstract
Glaucoma is the leading cause of irreversible blindness worldwide. Currently the only effective treatment for glaucoma is to reduce the intraocular pressure, which can halt the progression of the disease. Highlighting the importance of identifying individuals at risk of developing glaucoma and those with early-stage glaucoma will help patients receive treatment before sight loss. However, some cases of glaucoma do not have raised intraocular pressure. In fact, glaucoma is caused by a variety of different mechanisms and has a wide range of different subtypes. Understanding other risk factors, the underlying mechanisms, and the pathology of glaucoma might lead to novel treatments and treatment of underlying diseases. In this review we present the latest research into glaucoma including the genetics and molecular basis of the disease.
Collapse
Affiliation(s)
- Weiwei Wang
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital, Northwest University, Xi'an, 710004, Shaanxi Province, China.
| | - Huaizhou Wang
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| |
Collapse
|
2
|
Kolko M, Gazzard G, Baudouin C, Beier S, Brignole-Baudouin F, Cvenkel B, Fineide F, Hedengran A, Hommer A, Jespersen E, Messmer EM, Murthy R, Sullivan AG, Tatham AJ, Utheim TP, Vittrup M, Sullivan DA. Impact of glaucoma medications on the ocular surface and how ocular surface disease can influence glaucoma treatment. Ocul Surf 2023; 29:456-468. [PMID: 37302545 DOI: 10.1016/j.jtos.2023.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/13/2023]
Affiliation(s)
- Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | - Gus Gazzard
- Glaucoma Service, Moorfields Eye Hospital NHS Foundation Trust, London, UK; UCL Institute of Ophthalmology, London, UK; NIHR-Moorfields Biomedical Research Centre, London, UK
| | - Christophe Baudouin
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France; Université Paris-Saclay, Versailles-Saint-Quentin-en-Yvelines, Paris, France; Institut de la Vision, Sorbonne Université, Paris, France
| | - Sofie Beier
- Royal Danish Academy - Architecture, Design, Conservation, Copenhagen, Denmark
| | - Françoise Brignole-Baudouin
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France; Institut de la Vision, Sorbonne Université, Paris, France; Faculté de Pharmacie, Paris Cité université, Paris, France
| | - Barbara Cvenkel
- Department of Ophthalmology, University Medical Centre Ljubljana, Ljubljana, Slovenia; Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Fredrik Fineide
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Anne Hedengran
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | - Anton Hommer
- Department of Ophthalmology, HERA Hospital, Vienna, Austria
| | | | | | | | | | - Andrew J Tatham
- Princess Alexandra Eye Pavilion and Department of Ophthalmology, University of Edinburgh, Edinburgh, UK
| | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | | | | |
Collapse
|
3
|
Sun Y, Wirta D, Murahashi W, Mathur V, Sankaranarayanan S, Taylor LK, Yednock T, Fong DS, Goldberg JL. Safety and Target Engagement of C1q Inhibitor ANX007 in Neurodegenerative Eye Disease: Results from Phase 1 Studies in Glaucoma. OPHTHALMOLOGY SCIENCE 2023; 3:100290. [PMID: 37124168 PMCID: PMC10130689 DOI: 10.1016/j.xops.2023.100290] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/26/2023]
Abstract
Purpose Complement C1q, the initiating molecule of the classical complement cascade, is involved in synapse elimination and neuronal loss in neurodegenerative diseases including glaucoma. Here we report an evaluation of the safety, tolerability, and ocular pharmacokinetics (PK) and pharmacodynamics of intravitreal (IVT) injections of ANX007, an anti-C1q monoclonal antibody fragment that blocks activation of the classical complement cascade. Design An open-label, single-dose-escalation phase Ia study followed by a double-masked, randomized, sham-controlled, repeat-injection phase Ib study. Participants A total of 26 patients with primary open-angle glaucoma. Methods Nine patients with primary open-angle glaucoma (mean Humphrey visual field deviation between -3 and -18 decibels [dB]) were enrolled in phase Ia and received single doses of ANX007 (1.0 mg, n = 3; 2.5 mg, n = 3; or 5.0 mg, n = 3). Seventeen patients (mean Humphrey visual field deviation between -3 and -24 dB) were enrolled in phase Ib and randomized to 2 monthly doses of ANX007 (sham, n = 6; 2.5 mg ANX007, n = 6; or 5 mg ANX007, n = 5). Main Outcome Measures Safety and tolerability (including laboratory evaluation of urinalysis, complete blood count, and serum chemistries), ANX007 PK, target engagement, and immunogenicity. Results The mean age overall was 70 years in phase Ia and 68 years in phase Ib. In both studies, no serious adverse events were observed, no non-ocular treatment-emergent adverse events (TEAEs) attributable to study drug were reported, and ocular TEAEs were mild. Intraocular pressure returned to normal levels for all patients within 45 minutes of IVT injection. No clinically significant deviations in laboratory results were observed. In the phase Ib study, C1q in the aqueous humor was reduced to undetectable levels in both the 2.5 mg and 5 mg cohorts 4 weeks after the first ANX007 dose. Conclusions In these studies, single and repeat IVT ANX007 injections were well tolerated and demonstrated full target engagement 4 weeks after dosing with both low and high doses, supporting monthly or less-frequent dosing. Further investigation in neurodegenerative ocular diseases is warranted. Financial Disclosures Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Yang Sun
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California
| | - David Wirta
- Eye Research Foundation, Newport Beach, California
| | | | | | | | | | | | | | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California
- Correspondence: Jeffrey L. Goldberg, MD, PhD, Byers Eye Institute at Stanford, 2452 Watson Ct, Palo Alto, CA 94025.
| |
Collapse
|
4
|
Zanutigh V, Perrone LD, Gómez-Caride G, Perrone F, Valvecchia G, Logioco C. Success rate in micropulse diode laser treatment with regard to lens status, refractive errors, and glaucoma subtypes. Int Ophthalmol 2023:10.1007/s10792-023-02640-2. [PMID: 36715958 DOI: 10.1007/s10792-023-02640-2] [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: 08/20/2021] [Accepted: 01/19/2023] [Indexed: 01/31/2023]
Abstract
PURPOSE To evaluate the efficacy of micropulse transscleral cyclophotocoagulation (MP-TSCPC) considering different characteristics: glaucoma subtypes and lens status. METHODS A retrospective case-series study was designed to evaluate intraocular pressure (IOP), and the number of IOP-lowering medications, used by glaucoma patients treated with MP-TSCPC between 2016 and 2019. Cases had a follow-up period of 12 months. Achieving an IOP reduction higher than 20%, or the decrease of at least one IOP-lowering medication, was considered a successful outcome. The same population was analyzed by classifying them in two groups as: glaucoma subtypes and lens status. The baseline spherical equivalent (SE) was also calculated for considering association with the achieved IOP. RESULTS A total of 86 eyes were included. In most cases, IOP and IOP-lowering medications were decreased with a statistically significant difference (p < 0.0001), and all of them had a successful outcome. The percentage of IOP drop oscillated between 25.9% (open-angle glaucoma sub-group) and 37.5% (pseudoexfoliative glaucoma sub-group), 12 months after surgery. The difference between the groups was not statistically significant (p 0.20 and 0.32 for glaucoma subtypes and lens status, respectively). The Pearson's coefficient obtained was low for the SE and IOP association, at the 12 -month postoperative mark (- 0.009; p < 0.001). CONCLUSIONS The MP-TSCPC treatment was successful in decreasing IOP and IOP-lowering medications, in different glaucoma subtypes. Differences between groups (glaucoma subtypes, phakic and pseudophakic eyes) were not statistically significant. No association was found between the SE and the IOP achieved value after MS-TSCPC treatment.
Collapse
Affiliation(s)
- Virginia Zanutigh
- Centro de Ojos Quilmes, Humberto Primo, 298, Quilmes, Buenos Aires, Argentina.
| | | | - Gastón Gómez-Caride
- Centro de Ojos Quilmes, Humberto Primo, 298, Quilmes, Buenos Aires, Argentina
| | - Franco Perrone
- Centro de Ojos Quilmes, Humberto Primo, 298, Quilmes, Buenos Aires, Argentina
| | - Gerardo Valvecchia
- Centro de Ojos Quilmes, Humberto Primo, 298, Quilmes, Buenos Aires, Argentina
| | - Celina Logioco
- Centro de Ojos Quilmes, Humberto Primo, 298, Quilmes, Buenos Aires, Argentina
| |
Collapse
|
5
|
ANGPTL7, a therapeutic target for increased intraocular pressure and glaucoma. Commun Biol 2022; 5:1051. [PMID: 36192519 PMCID: PMC9529959 DOI: 10.1038/s42003-022-03932-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/01/2022] [Indexed: 12/03/2022] Open
Abstract
Glaucoma is a leading cause of blindness. Current glaucoma medications work by lowering intraocular pressure (IOP), a risk factor for glaucoma, but most treatments do not directly target the pathological changes leading to increased IOP, which can manifest as medication resistance as disease progresses. To identify physiological modulators of IOP, we performed genome- and exome-wide association analysis in >129,000 individuals with IOP measurements and extended these findings to an analysis of glaucoma risk. We report the identification and functional characterization of rare coding variants (including loss-of-function variants) in ANGPTL7 associated with reduction in IOP and glaucoma protection. We validated the human genetics findings in mice by establishing that Angptl7 knockout mice have lower (~2 mmHg) basal IOP compared to wild-type, with a trend towards lower IOP also in heterozygotes. Conversely, increasing murine Angptl7 levels via injection into mouse eyes increases the IOP. We also show that acute Angptl7 silencing in adult mice lowers the IOP (~2-4 mmHg), reproducing the observations in knockout mice. Collectively, our data suggest that ANGPTL7 is important for IOP homeostasis and is amenable to therapeutic modulation to help maintain a healthy IOP that can prevent onset or slow the progression of glaucoma.
Collapse
|
6
|
Bastia E, Sgambellone S, Lucarini L, Provensi G, Brambilla S, Galli C, Almirante N, Impagnatiello F. NCX 470 Restores Ocular Hemodynamics and Retinal Cell Physiology After ET-1-Induced Ischemia/Reperfusion Injury of Optic Nerve and Retina in Rabbits. J Ocul Pharmacol Ther 2022; 38:496-504. [PMID: 35787180 DOI: 10.1089/jop.2022.0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Purpose: Determine whether NCX 470, a nitric oxide (NO)-donating bimatoprost with clinically demonstrated intraocular pressure (IOP)-lowering effects, improves ocular hemodynamics and retinal physiology. Methods: Endothelin-1 (ET-1)-induced ischemia/reperfusion model in New Zealand white rabbits was used. ET-1 was injected next to the optic nerve twice/week (Monday and Thursday) for 6 weeks. Starting on week 3, animals received NCX 470 (0.1% bid, 6 days/week Monday-Saturday) or vehicle until the end of ET-1 treatment. IOP, ophthalmic artery resistive index (OA-RI) and retina physiology (electroretinogram, ERG) were determined before dosing and at different times post-dosing. All measurements were taken on Mondays before the AM daily dosing (36 h treatment-free). Finally, oxidative stress markers were determined in dissected retina and iris/ciliary body of treated eyes. Results: Injection of ET-1 progressively increased IOP (20.7 ± 0.6, 24.9 ± 1.2, and 27.0 ± 0.6 mmHg at baseline, week 2 and 6, respectively) and OA-RI (0.30 ± 0.02, 0.39 ± 0.02, and 0.42 ± 0.03 at baseline, week 2 and 6, respectively) and reduced rods and/or cones response as indicated by changes in ERG amplitudes under different stimulating conditions. NCX 470 re-established baseline IOP (21.8 ± 1.0 mmHg), OA-RI (0.33 ± 0.02), and ERG amplitude by week 6 (mostly rod response, 0.01Dark_AVeh_6week = 32.2 ± 3.0 μV and 0.01Dark_ANCX470_6week 44.3 ± 4.5 μV; mostly cone response, 3.0Dark_AVeh_6week = 87.6 ± 10.1 μV and 3.0Dark_ANCX470_6week = 122.8 ± 11.4 μV; combined rod/cone response, 3.0Light_AVeh_6week = 49.8 ± 6.5 μV and 3.0Light_ANCX470_6week = 64.2 ± 6.8 μV). NCX 470 also reversed ET-1-induced changes in glutathione and manganese superoxide dismutase (oxidative stress markers) in retina and iris/ciliary body. Conclusions: Repeated ocular topical dosing with NCX 470 reverses ET-1-induced changes in IOP, OA-RI, and ERG suggesting improved ocular hemodynamics and retinal physiology likely independently from its demonstrated IOP-lowering effect.
Collapse
Affiliation(s)
| | - Silvia Sgambellone
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy
| | - Laura Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy
| | - Gustavo Provensi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy
| | | | | | | | | |
Collapse
|
7
|
Fea AM, Cattel F, Gandolfi S, Buseghin G, Furneri G, Costagliola C. Cost-utility analysis of trabecular micro-bypass stents (TBS) in patients with mild-to-moderate open-angle Glaucoma in Italy. BMC Health Serv Res 2021; 21:824. [PMID: 34399759 PMCID: PMC8369731 DOI: 10.1186/s12913-021-06862-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/09/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Glaucoma is a disease characterized by progressive damage of the optic nerve. Several therapeutic options are available to lower intraocular pressure (IOP). In primary open-angle glaucoma (POAG) patients with inadequate IOP control (or controlled with multiple medical therapies or for whom medical therapy is contraindicated), the implantation of micro-invasive glaucoma surgery devices (MIGS) and concomitant cataract surgery has proved to be more effective in reducing intraocular pressure (IOP), as compared to cataract surgery alone. The objective of this study was to assess the cost-effectiveness of iStent inject® device with concurrent cataract surgery vs. cataract surgery alone, in patients with mild-to-moderate POAG, adopting the Italian National Health Service (NHS) perspective. METHODS Simulation of outcomes and costs was undertaken using a Markov model with 4 health states and one-month cycles, that is used to simulate the prognosis of these patients. Efficacy data were obtained from the randomized clinical trial (RCT). A lifetime horizon was adopted in the analysis. A discount rate of 3.5% was applied to both costs and effects. The Italian National Healthcare Service (NHS) perspective was considered, therefore only healthcare direct costs (acquisition of main interventions and subsequent procedures; medications; monitoring and follow-up; adverse events). Model robustness was tested through sensitivity analyses. RESULTS Results of the base-case analysis showed that the total lifetime costs were higher in the iStent inject® + concurrent cataract surgery, compared with the cataract surgery alone group (€8368.51 vs. €7134.71 respectively). iStent inject® + concurrent cataract surgery was cost-effective vs. cataract surgery alone, with an incremental cost-effectiveness ratio of €13,037.01 per quality-adjusted life year (QALY) gained. Both one-way deterministic and probabilistic sensitivity analyses confirmed robustness of base-case results. The cost-effectiveness accessibility curve (CEAC) showed that iStent inject® + cataract surgery would have a 98% probability of being cost-effective, compared to cataract surgery alone, when the willingness to pay (WTP) is equal to €50,000 per QALY gained. CONCLUSIONS The results of the cost-utility analysis confirm that iStent inject® + cataract surgery is a cost-effective option for the treatment of patients affected by mild-to-moderate POAG, compared with cataract surgery alone, when evaluated from the Italian NHS perspective.
Collapse
Affiliation(s)
| | - Francesco Cattel
- Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, City of Science and Health Molinette, Hospital Pharmacy, Turin, Italy
| | - Stefano Gandolfi
- Ophthalmology Unit, Department of Biological, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| | | | | | - Ciro Costagliola
- Department of Medicine & Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| |
Collapse
|
8
|
Sit AJ, Gupta D, Kazemi A, McKee H, Challa P, Liu KC, Lopez J, Kopczynski C, Heah T. Netarsudil Improves Trabecular Outflow Facility in Patients with Primary Open Angle Glaucoma or Ocular Hypertension: A Phase 2 Study. Am J Ophthalmol 2021; 226:262-269. [PMID: 33524367 DOI: 10.1016/j.ajo.2021.01.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE Intraocular pressure (IOP) reduction is key to controlling primary open angle glaucoma (POAG). Pharmacotherapies for POAG or ocular hypertension (OHT) commonly lower IOP by increasing uveoscleral outflow or decreasing aqueous humor production. Netarsudil (Rhopressa), a Rho kinase inhibitor, reduces IOP by improving trabecular outflow facility, which is reduced in POAG. We investigated the effects of netarsudil on aqueous humor dynamics in patients with POAG or OHT. DESIGN Double-masked, randomized, vehicle-controlled, Phase 2 trial. METHODS Netarsudil 0.02% was instilled in 1 eye and vehicle into the contralateral eye of 20 patients once daily in the morning for 7 days. The primary endpoint was change in mean diurnal outflow facility on day 8 versus that on day 1 (baseline). Outflow facility was measured by using Schiøtz tonography, IOP by pneumotonometry, and episcleral venous pressure (EVP) by automated venomanometry. RESULTS Eighteen patients (90%) completed the study. Mean diurnal outflow facility increased 0.039 versus 0.007 µL/min/mm Hg from baseline in the netarsudil- and the vehicle-treated groups, respectively (P < .001 vs. baseline for netarsudil), a treatment difference of 0.03 µL/min/mm Hg (P ≤ .001). Mean diurnal IOP change from baseline at day 8 was -4.52 mm Hg for netarsudil versus -0.98 mm Hg for vehicle, a treatment difference of -3.54 mm Hg (P < .0001). Mean diurnal EVP change from baseline was -0.79 mm Hg in the netarsudil-treated group versus 0.10 mm Hg for vehicle, a treatment difference of -0.89 mm Hg (P < .001). All patients reporting an adverse event reported conjunctival hyperemia of mild or moderate severity. CONCLUSIONS Netarsudil acts on the conventional outflow pathway, both proximal and distal, to significantly reduce IOP in POAG and OHT by improving trabecular outflow facility and decreasing EVP.
Collapse
Affiliation(s)
- Arthur J Sit
- From the Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA.
| | - Divakar Gupta
- Department of Ophthalmology, Duke University, Durham, North Carolina, USA
| | - Arash Kazemi
- From the Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hayley McKee
- Aerie Pharmaceuticals, Inc., Durham, North Carolina, USA
| | - Pratap Challa
- Department of Ophthalmology, Duke University, Durham, North Carolina, USA
| | - Katy C Liu
- Department of Ophthalmology, Duke University, Durham, North Carolina, USA
| | - Jae Lopez
- Aerie Pharmaceuticals, Inc., Durham, North Carolina, USA
| | | | - Theresa Heah
- Aerie Pharmaceuticals, Inc., Durham, North Carolina, USA
| |
Collapse
|