Single Administration of Intracameral Bimatoprost Implant 10 µg in Patients with Open-Angle Glaucoma or Ocular Hypertension

Felipe A Medeiros, Arsham Sheybani, Manjool M Shah, Marcos Rivas, Zhanying Bai, Erica Werts, Iqbal I K Ahmed, E Randy Craven, Felipe A Medeiros, Arsham Sheybani, Manjool M Shah, Marcos Rivas, Zhanying Bai, Erica Werts, Iqbal I K Ahmed, E Randy Craven

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.

Keywords: Adherence; Drug delivery system; Drug implant; Glaucoma; Prostaglandin analog.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
IOP in study eyes after a single administration of 10-µg bimatoprost implant or topical timolol maleate 0.5% BID through week 15 in the pooled ARTEMIS phase 3 studies. The analysis used observed values for eyes that had not received any rescue IOP-lowering treatment. a Mean ± SD IOP. b Postbaseline IOP was compared between groups at hours 0 and 2 at weeks 2, 6, 12, and 15 using a mixed-effects model for repeated measures. Least-squares estimates of mean ± SE IOP at postbaseline time points, the differences between groups (bimatoprost implant minus timolol), and the 95% CIs of the between-group differences were derived from the model. Baseline IOP is mean ± SE calculated in a separate analysis. BID twice daily, CI confidence interval, IOP intraocular pressure, SD standard deviation, SE standard error
Fig. 2
Fig. 2
Responder rates for a 10-µg bimatoprost implant vs topical timolol maleate 0.5% BID in the pooled ARTEMIS phase 3 studies and b 10-µg bimatoprost implant vs topical bimatoprost 0.03% QD in the phase 1/2 study. BID twice daily, IOP intraocular pressure, QD once daily
Fig. 3
Fig. 3
IOP in non-rescued eyes after a single administration of 10-µg bimatoprost implant or once-daily topical bimatoprost 0.03% through month 24 in the phase 1/2 study. The analysis used observed values for eyes that had not received any rescue IOP-lowering treatment or a second administration of implant. Values shows are mean ± SD. The differences between groups (study eye minus fellow eye) and 95% CIs of the differences were calculated using paired t tests. CI confidence interval, H hour, IOP intraocular pressure, SD standard deviation
Fig. 4
Fig. 4
Kaplan–Meier survival analysis of time to initial use of any additional IOP-lowering treatment (rescue topical medication or a second administration of implant) after a single administration of bimatoprost implant 10 µg on day 1 in the phase 1/2 study. The estimated median time to use of additional IOP-lowering treatment was 273 days. At 720 days, the number of patients at risk was 3 rather than 5, because 2 patients had already completed the study without rescue or re-treatment. IOP intraocular pressure
Fig. 5
Fig. 5
Proportion of study eyes with loss in CECD from baseline to week 12 after administration of a 10-µg bimatoprost implant or beginning topical timolol BID treatment on day 1 in the pooled ARTEMIS phase 3 studies. BID twice daily, CECD central corneal endothelial cell density
Fig. 6
Fig. 6
Mean ± SD CECD at baseline and during treatment in the phase 1/2 study. Study eyes received a single administration of bimatoprost implant 10 µg, and fellow eyes were treated with topical bimatoprost 0.03% QD. Eyes were censored from the analysis at the time of implant re-treatment but could have received topical rescue IOP-lowering medication and remained in the analysis. The differences between groups (study eye minus fellow eye) and 95% CIs of the differences were calculated using paired t tests. BL baseline, CECD central corneal endothelial cell density, CI confidence interval, IOP intraocular pressure, M month, QD once daily, SD standard deviation, W week

References

    1. Olthoff CMG, Schouten JSAG, van de Borne BW, Webers CAB. Noncompliance with ocular hypotensive treatment in patients with glaucoma or ocular hypertension an evidence-based review. Ophthalmology. 2005;112(6):953–961. doi: 10.1016/j.ophtha.2004.12.035.
    1. Yeaw J, Benner JS, Walt JG, Sian S, Smith DB. Comparing adherence and persistence across 6 chronic medication classes. J Manag Care Pharm. 2009;15(9):728–740. doi: 10.18553/jmcp.2009.15.9.728.
    1. Newman-Casey PA, Robin AL, Blachley T, et al. The most common barriers to glaucoma medication adherence: a cross-sectional survey. Ophthalmology. 2015;122(7):1308–1316. doi: 10.1016/j.ophtha.2015.03.026.
    1. Gomes BF, Paredes AF, Madeira N, Moraes HV, Jr, Santhiago MR. Assessment of eye drop instillation technique in glaucoma patients. Arq Bras Oftalmol. 2017;80(4):238–241. doi: 10.5935/0004-2749.20170058.
    1. Okeke CO, Quigley HA, Jampel HD, et al. Adherence with topical glaucoma medication monitored electronically the Travatan Dosing Aid study. Ophthalmology. 2009;116(2):191–199. doi: 10.1016/j.ophtha.2008.09.004.
    1. Schwartz GF, Hollander DA, Williams JM. Evaluation of eye drop administration technique in patients with glaucoma or ocular hypertension. Curr Med Res Opin. 2013;29(11):1515–1522. doi: 10.1185/03007995.2013.833898.
    1. Robin AL, Muir KW. Medication adherence in patients with ocular hypertension or glaucoma. Expert Rev Ophthalmol. 2019;14:199–210. doi: 10.1080/17469899.2019.1635456.
    1. Rossi GC, Pasinetti GM, Scudeller L, Radaelli R, Bianchi PE. Do adherence rates and glaucomatous visual field progression correlate? Eur J Ophthalmol. 2011;21(4):410–414. doi: 10.5301/EJO.2010.6112.
    1. Sleath B, Blalock S, Covert D, et al. The relationship between glaucoma medication adherence, eye drop technique, and visual field defect severity. Ophthalmology. 2011;118(12):2398–2402. doi: 10.1016/j.ophtha.2011.05.013.
    1. Newman-Casey PA, Niziol LM, Gillespie BW, Janz NK, Lichter PR, Musch DC. The association between medication adherence and visual field progression in the Collaborative Initial Glaucoma Treatment Study. Ophthalmology. 2020;127(4):477–483. doi: 10.1016/j.ophtha.2019.10.022.
    1. Gedde SJ, Vinod K, Wright MM, et al. Primary open-angle glaucoma Preferred Practice Pattern®. Ophthalmology. 2021;128(1):P71–150.
    1. Lee SS, Hughes P, Ross AD, Robinson MR. Biodegradable implants for sustained drug release in the eye. Pharm Res. 2010;27(10):2043–2053. doi: 10.1007/s11095-010-0159-x.
    1. Medeiros FA, Walters TR, Kolko M, et al. Phase 3, randomized, 20-month study of bimatoprost implant in open-angle glaucoma and ocular hypertension (ARTEMIS 1). Ophthalmology. 2020;127(12):1627–41.
    1. Seal JR, Robinson MR, Burke J, Bejanian M, Coote M, Attar M. Intracameral sustained-release bimatoprost implant delivers bimatoprost to target tissues with reduced drug exposure to off-target tissues. J Ocul Pharmacol Ther. 2019;35(1):50–57. doi: 10.1089/jop.2018.0067.
    1. Craven ER, Walters T, Christie WC, et al. 24-Month phase I/II clinical trial of Bimatoprost sustained-release implant (Bimatoprost SR) in glaucoma patients. Drugs. 2020;80(2):167–79. doi: 10.1007/s40265-019-01248-0.
    1. Lewis RA, Christie WC, Day DG, et al. Bimatoprost sustained-release implants for glaucoma therapy: 6-month results from a phase I/II clinical trial. Am J Ophthalmol. 2017;175:137–47. doi: 10.1016/j.ajo.2016.11.020.
    1. Bacharach J, Tatham A, Ferguson G, et al. 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(17):2017–33.
    1. Grippo TM, Liu JH, Zebardast N, Arnold TB, Moore GH, Weinreb RN. Twenty-four-hour pattern of intraocular pressure in untreated patients with ocular hypertension. Invest Ophthalmol Vis Sci. 2013;54(1):512–517. doi: 10.1167/iovs.12-10709.
    1. Allergan. Durysta (bimatoprost implant) prescribing information. 2020. . Accessed 19 Jan 2022.
    1. Khawaja AP, Campbell JH, Kirby N, et al. Real-world outcomes of selective laser trabeculoplasty in the United Kingdom. Ophthalmology. 2020;127(6):748–57.
    1. Weinreb RN, Robinson MR, Dibas M, Stamer WD. Matrix metalloproteinases and glaucoma treatment. J Ocul Pharmacol Ther. 2020;36(4):208–228. doi: 10.1089/jop.2019.0146.
    1. McCarey BE, Edelhauser HF, Lynn MJ. Review of corneal endothelial specular microscopy for FDA clinical trials of refractive procedures, surgical devices, and new intraocular drugs and solutions. Cornea. 2008;27(1):1–16. doi: 10.1097/ICO.0b013e31815892da.

Source: PubMed

3
Subskrybuj