ONSET-1 Phase 2b Randomized Trial to Evaluate the Safety and Efficacy of OC-01 (Varenicline Solution) Nasal Spray on Signs and Symptoms of Dry Eye Disease

David Wirta, Gail L Torkildsen, Blair Boehmer, David A Hollander, Edward Bendert, Lijuan Zeng, Michael Ackermann, Jeffrey Nau, David Wirta, Gail L Torkildsen, Blair Boehmer, David A Hollander, Edward Bendert, Lijuan Zeng, Michael Ackermann, Jeffrey Nau

Abstract

Purpose: The purpose of this trial was to evaluate the safety and efficacy of OC-01 (varenicline solution), a nicotinic acetylcholine receptor agonist nasal spray, on signs and symptoms of dry eye disease.

Methods: A phase 2b, multicenter, randomized, double-masked, vehicle-controlled trial (ONSET-1; NCT03636061) was performed. Patients were aged 22 years or older with a physician's diagnosis of dry eye disease and previous use of artificial tears were randomized 1:1:1:1 to control (vehicle nasal spray twice daily [BID]), OC-01 0.006 mg BID, OC-01 0.03 mg BID, and OC-01 0.06 mg BID. The primary end point was the change in the anesthetized Schirmer test score from baseline to day 28 in the study eye. The secondary end points included the change in the eye dryness score from baseline to day 28.

Results: One hundred eighty-two patients were randomized. After 28 days, patients who received OC-01 0.03 or 0.06 mg showed a statistically significant improvement in tear film production relative to vehicle, with least squares mean differences from vehicle of 7.7 mm [95% confidence interval, 3.8-11.7; P < 0.001] with OC-01 0.03 mg and 7.5 mm (95% confidence interval, 3.4-11.6; P < 0.001) with OC-01 0.06 mg. Patients receiving OC-01 0.03 mg showed a significant reduction in the eye dryness score by day 28 versus vehicle (P = 0.021); those receiving the OC-01 0.06 mg dose showed a nonsignificant reduction versus vehicle. OC-01 administration was associated with sneezing (62%-84%) and cough (9%-25%); these were transient and predominantly mild in severity.

Conclusions: OC-01 nasal spray administered BID at 0.03 and 0.06 mg resulted in significant improvements in signs and symptoms of dry eye disease, was well tolerated, and warrants further clinical investigation.

Conflict of interest statement

D. Wirta and G. L. Torkildsen have received research grant support from Oyster Point Pharma, Inc. D. A. Hollander is an employee of Ora, Inc. E. Bendert was an employee at Statistics Collaborative, Inc at the time the study was conducted. L. Zeng is a current employee at Statistics Collaborative, Inc. M. Ackermann is a board member at Oyster Point Pharma, Inc. J. Nau is an employee at and shareholder of Oyster Point Pharma, Inc. The remaining author has no conflicts of interest to disclose.

Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc.

Figures

FIGURE 1.
FIGURE 1.
CONSORT flow diagram. AE, adverse event; OSDI, Ocular Surface Disease Index; PI, principal investigator.
FIGURE 2.
FIGURE 2.
A, LS mean change from baseline to day 28 in STS. B, The LS mean change from baseline to day 28 in STS with missing data imputed using last available data. C, The percentage of patients with at least a 10-mm change in STS at day 28 versus baseline (intent-to-treat population) with missing data imputed using last available data. All comparisons made with the control group. No formal statistical comparisons were performed for the 0.006 mg dose versus vehicle; P values for this dose were assessed post hoc and represent nominal values. Error bars indicate CIs. The results in A are derived from a prespecified analysis and B and C are from a post hoc analysis.
FIGURE 3.
FIGURE 3.
A, Mean reduction in EDS from baseline to day 28. B, The LS mean reduction from baseline to day 28 with missing data imputed using last available data. C, The LS mean reduction in the controlled adverse environment at day 21. All comparisons made with the control group. No formal statistical comparisons were performed for the 0.006 mg dose versus vehicle; P values for this dose were assessed post hoc and represent nominal values. Error bars indicate CIs. The results in A are derived from a prespecified analysis and B and C are from post hoc analyses.

References

    1. Friedman NJ, Butron K, Robledo N, et al. . A nonrandomized, open-label study to evaluate the effect of nasal stimulation on tear production in subjects with dry eye disease. Clin Ophthalmol. 2016;10:795–804.
    1. Craig JP, Nichols KK, Akpek EK, et al. . TFOS DEWS II definition and classification report. Ocul Surf. 2017;15:276–283.
    1. Paulsen AJ, Cruickshanks KJ, Fischer ME, et al. . Dry eye in the Beaver Dam Offspring Study: prevalence, risk factors, and health-related quality of life. Am J Ophthalmol. 2014;157:799–806.
    1. Bhavsar AS, Bhavsar SG, Jain SM. A review on recent advances in dry eye: pathogenesis and management. Oman J Ophthalmol. 2011;4:50–56.
    1. Lemp MA. Advances in understanding and managing dry eye disease. Am J Ophthalmol. 2008;146:350–356.
    1. Willcox MDP, Argüeso P, Georgiev GA, et al. . TFOS DEWS II tear film report. Ocul Surf. 2017;15:366–403.
    1. Schicht M, Garreis F, Hartjen N, et al. . SFTA3—a novel surfactant protein of the ocular surface and its role in corneal wound healing and tear film surface tension. Sci Rep. 2018;8:9791.
    1. Klenkler B, Sheardown H, Jones L. Growth factors in the tear film: role in tissue maintenance, wound healing, and ocular pathology. Ocul Surf. 2007;5:228–239.
    1. Suwan-apichon O, Rizen M, Rangsin R, et al. . Botulinum toxin B-induced mouse model of keratoconjunctivitis sicca. Invest Ophthalmol Vis Sci. 2006;47:133–139.
    1. Zhu L, Shen J, Zhang C, et al. . Inflammatory cytokine expression on the ocular surface in the botulium toxin B induced murine dry eye model. Mol Vis. 2009;15:250–258.
    1. Flanagan JL, Willcox MD. Role of lactoferrin in the tear film. Biochimie. 2009;91:35–43.
    1. McKown RL, Wang N, Raab RW, et al. . Lacritin and other new proteins of the lacrimal functional unit. Exp Eye Res. 2009;88:848–858.
    1. Wang J, Wang N, Xie J, et al. . Restricted epithelial proliferation by lacritin via PKCα-dependent NFAT and mTOR pathways. J Cell Biol. 2006;174:689–700.
    1. Dartt DA. Neural regulation of lacrimal gland secretory processes: relevance in dry eye diseases. Prog Retin Eye Res. 2009;28:155–177.
    1. Alimohammadi H, Silver WL. Evidence for nicotinic acetylcholine receptors on nasal trigeminal nerve endings of the rat. Chem Senses. 2000;25:61–66.
    1. Gupta A, Heigle T, Pflugfelder SC. Nasolacrimal stimulation of aqueous tear production. Cornea. 1997;16:645–648.
    1. Dieckmann G, Jamali A, Pondelis N, et al. . In vivo confocal microscopy demonstrates intranasal neurostimulation-induced goblet cell alterations in patients with dry eye disease. Invest Ophthalmol Vis Sci. 2017;58:2694.
    1. Green KB, Kamat M, Franke M, et al. . Tear total lipid concentration in patients with dry eye following intranasal neurostimulation. Invest Ophthalmol Vis Sci. 2017;58:2693.
    1. Gumus K, Schuetzle KL, Pflugfelder SC. Randomized controlled crossover trial comparing the impact of sham or intranasal tear neurostimulation on conjunctival goblet cell degranulation. Am J Ophthalmol. 2017;177:159–168.
    1. Pondelis NJ, Dieckmann G, Kataguiri P, et al. . Intranasal neurostimulator induces morphological changes in meibomian glands in patients with dry eye disease. Invest Ophthalmol Vis Sci. 2017;58:2235.
    1. Sheppard JD, Torkildsen GL, Geffin JA, et al. . Characterization of tear production in subjects with dry eye disease during intranasal tear neurostimulation: results from two pivotal clinical trials. Ocul Surf. 2019;17:142–150.
    1. Woodward A, Senchyna M, Franke M, et al. . Effect of intranasal neurostimulation on tear protein content in patient with dry eye. Invest Ophthalmol Vis Sci. 2017;58:2673.
    1. O'Neil EC, Henderson M, Massaro-Giordano M, et al. . Advances in dry eye disease treatment. Curr Opin Ophthalmol. 2019;30:166–178.
    1. Mihalak KB, Carroll FI, Luetje CW. Varenicline is a partial agonist at alpha4beta2 and a full agonist at alpha7 neuronal nicotinic receptors. Mol Pharmacol. 2006;70:801–805.
    1. Li N, Deng X-G, He MF. Comparison of the Schirmer I test with and without topical anesthesia for diagnosing dry eye. Int J Ophthalmol. 2012;5:478–481.
    1. Sheppard JD, Torkildsen GL, Lonsdale JD, et al. . Lifitegrast ophthalmic solution 5.0% for treatment of dry eye disease: results of the OPUS-1 phase 3 study. Ophthalmology. 2014;121:475–483.
    1. Tauber J, Karpecki P, Latkany R, et al. ; OPUS-2 Investigators. Lifitegrast ophthalmic solution 5.0% versus placebo for treatment of dry eye disease: results of the randomized phase III OPUS-2 study. Ophthalmology. 2015;122:2423–2431.
    1. Ousler GW, Gomes PJ, Welch D, et al. . Methodologies for the study of ocular surface disease. Ocul Surf. 2005;3:143–154.
    1. Ousler GW, III, Rimmer D, Smith LM, et al. . Use of the controlled adverse environment (CAE) in clinical research: a review. Ophthalmol Ther. 2017;6:263–276.
    1. Pattar GR, Jerkins G, Evans DG, et al. . Symptom improvement in dry eye subjects following intranasal tear neurostimulation: results of two studies utilizing a controlled adverse environment. Ocul Surf. 2020;18:249–257.
    1. Batsel HL, Lines AJ. Neural mechanisms of sneeze. Am J Physiol. 1975;229:770–776.
    1. Songu M, Cingi C. Sneeze reflex: facts and fiction. Ther Adv Respir Dis. 2009;3:131–141.
    1. Hydén D, Arlinger S. On the sneeze-reflex and its control. Rhinology. 2007;45:218–219.
    1. FDA Advisory Committees Recommend to Remove Boxed Warning in Labeling for Pfizer's Smoking Cessation Therapy, CHANTIX® (Varenicline). New York, NY: Pfizer; 2016. Available at: . Accessed February 26, 2020.
    1. Torkildsen G, Brujic M, Cooper MS, et al. . Evaluation of a new artificial tear formulation for the management of tear film stability and visual function in patients with dry eye. Clin Ophthalmol. 2017;11:1883–1889.
    1. Campling BG, Kuryatov A, Lindstrom J. Acute activation, desensitization and smoldering activation of human acetylcholine receptors. PLoS One. 2013;8:e79653.
    1. Ghori MU, Mahdi MH, Smith AM, et al. . Nasal drug delivery systems: an overview. Am J Pharmacol Sci. 2015;3:110–119.
    1. Silver WL, Finger TE. The anatomical and electrophysiological basis of peripheral nasal trigeminal chemoreception. Ann N Y Acad Sci. 2009;1170:202–205.
    1. Hofmann T, Gugatschga M, Koidl B, et al. . Influence of preservatives and topical steroids on ciliary beat frequency in vitro. Arch Otolaryngol Head Neck Surg. 2004;130:440–445.

Source: PubMed

3
Abonnere