Discovery and Preclinical Development of Netarsudil, a Novel Ocular Hypotensive Agent for the Treatment of Glaucoma

Cheng-Wen Lin, Bryan Sherman, Lori A Moore, Carmen L Laethem, Da-Wen Lu, Padmanabhan P Pattabiraman, Ponugoti Vasantha Rao, Mitchell A deLong, Casey C Kopczynski, Cheng-Wen Lin, Bryan Sherman, Lori A Moore, Carmen L Laethem, Da-Wen Lu, Padmanabhan P Pattabiraman, Ponugoti Vasantha Rao, Mitchell A deLong, Casey C Kopczynski

Abstract

Purpose: Rho-associated protein kinase (ROCK) inhibitors lower intraocular pressure (IOP) by increasing aqueous outflow through the trabecular meshwork (TM). The preclinical characterization of netarsudil, a new ROCK/norepinephrine transporter (NET) inhibitor currently in clinical development, is presented herein.

Methods: The kinase inhibitory activity of netarsudil was compared to its esterase metabolite, netarsudil-M1, and 3 other ROCK inhibitors using a commercially available kinase assay kit. Disruption of actin stress fibers was measured in primary porcine TM cells and disruption of focal adhesions in transformed human TM (HTM) cells. Induction of fibrosis markers after exposure to transforming growth factor-β2 (TGF-β2) was conducted in primary HTM cells. Ocular hypotensive activity and tolerability of topical formulations were evaluated in normotensive Dutch Belted rabbits and Formosan Rock monkeys. In vitro corneal metabolism assays were conducted using dog, pig, rabbit, monkey, and human corneas. In vivo ocular pharmacokinetics was studied in Dutch Belted rabbits.

Results: Netarsudil inhibited kinases ROCK1 and ROCK2 with a Ki of 1 nM each, disrupted actin stress fibers and focal adhesions in TM cells with IC50s of 79 and 16 nM, respectively, and blocked the profibrotic effects of TGF-β2 in HTM cells. Netarsudil produced large reductions in IOP in rabbits and monkeys that were sustained for at least 24 h after once daily dosing, with transient, mild hyperemia observed as the only adverse effect.

Conclusion: Netarsudil is a novel ROCK/NET inhibitor with high potency in biochemical and cell-based assays, an ability to produce large and durable IOP reductions in animal models, and favorable pharmacokinetic and ocular tolerability profiles.

Keywords: Rho kinase; glaucoma; intraocular pressure; netarsudil; trabecular meshwork.

Conflict of interest statement

C.-W.L., B.S., L.A.M., C.L.L., M.A.d.L., and C.C.K. are employees of and stockholders in Aerie Pharmaceuticals, Inc. D.-W.L. and P.V.R. received research funding from Aerie Pharmaceuticals.

Figures

FIG. 1.
FIG. 1.
Structures of test compounds. NETi, norepinephrine transporter inhibitor; ROCKi, Rho-associated protein kinase inhibitor.
FIG. 2.
FIG. 2.
Disruption of actin stress fibers and focal adhesions by netarsudil versus other ROCK inhibitors. (A) Netarsudil dose-response in actin stress fiber assay. Primary PTM cells were incubated for 6 h in the presence of 0, 0.015, 0.138, or 1.2 μM netarsudil then fixed and stained with Alexa Fluor-488 phalloidin and Hoechst 33342 to reveal actin fibers and nuclei, respectively. Top panel: fluorescence images of stained cells. Bottom panel: False color images created by an automated, custom algorithm to identify stress fibers and calculate mean stress fiber length. (B) Netarsudil dose-response in focal adhesion assay. Immortalized HTM cells (TM-1) were incubated for 6 h in the presence of 0, 0.015, 0.138, or 1.2 μM netarsudil then fixed and stained with mouse anti-paxillin antibody/Alexa Fluor®488 goat-anti-mouse IgG and Hoechst 33342 to reveal focal adhesions and nuclei, respectively. Top panel: fluorescence images of stained cells. Bottom panel: False color images created by an automated, custom algorithm to identify focal adhesions and calculate the mean number of focal adhesions per cell. (C) Dose–response curves (n = 4) for netarsudil, netarsudil-M1, AR-12286, Y-27632, and fasudil in the PTM actin stress fiber length assay. Mean stress fiber length is presented as a percentage of the mean length of stress fibers measured in untreated control cells. (D) Dose–response curves (n = 4) for netarsudil, netarsudil-M1, AR-12286, Y-27632, and fasudil in the HTM focal adhesion assay. Mean number of focal adhesions per cell is presented as a percentage of the number of focal adhesions per cell measured in untreated control cells. HTM, human trabecular meshwork; PTM, porcine trabecular meshwork; ROCK, Rho-associated protein kinase.
FIG. 3.
FIG. 3.
Netarsudil blocks the profibrotic effects of TGF-β on HTM cells. Serum-starved primary HTM cells incubated for 24 h in the presence of either vehicle, 8 ng/mL human TGF-β2, 500 nM netarsudil, or 8 ng/mL TGF-β2 plus 500 nM netarsudil were fixed and stained for the fibrogenic markers α-SMA, fibroblast-specific protein 1 (FSP1), and Collagen 1A. α-SMA, α-smooth muscle actin; HTM, human trabecular meshwork; TGF-β2, transforming growth factor-β2.
FIG. 4.
FIG. 4.
Netarsudil dose-dependent lowering of IOP in rabbits and monkeys. Formulations containing 0.005% (rabbit only), 0.01%, 0.02%, or 0.04% netarsudil were administered once daily (AM) to 1 eye of each animal for 3 days, with the fellow untreated eye serving as the control. IOP was determined for both eyes before test article administration (time 0) and at times 1, 2, 4, 8, and 24 h (rabbits) or 4, 8, and 24 h (monkeys) after each morning dose on Day 1 and 3. (A) Changes in IOP for the treated eye relative to the untreated contralateral eye in Dutch Belted rabbits (n = 10/group). IOP reductions were statistically significant (P < 0.05) at all postdose time points. (B) Change in IOP in the netarsudil-treated eye relative to the untreated contralateral eye in Formosan Rock monkeys (n = 6/group). IOP reductions were statistically significant (P < 0.05) at all postdose time points. IOP, intraocular pressure.
FIG. 5.
FIG. 5.
IOP-lowering effect of netarsudil versus AR-12286 in rabbits and monkeys. Netarsudil 0.04% or AR-12286 0.5% was administered once daily (AM) to 1 eye of each animal for 3 days, with the fellow untreated eye serving as the control. IOP was determined for both eyes before test article administration (time 0) and at times 1, 2, 4, 8, and 24 h (rabbits) or 4, 8, and 24 h (monkeys) after each morning dose on Day 1 and 3. (A) Change in IOP in the treated eye relative to the untreated contralateral eye in Dutch Belted rabbits (n = 12/group). For netarsudil 0.04% and AR-12286 0.5%, IOP reductions were statistically significant (P < 0.01) at all postdose time points. (B) Change in IOP in the treated eye relative to the untreated contralateral eye in Formosan Rock monkeys (n = 6/group). For netarsudil 0.04% and AR-12286 0.5%, IOP reductions were statistically significant (P < 0.01) at all postdose time points. IOP, intraocular pressure.
FIG. 6.
FIG. 6.
Metabolism of netarsudil by ocular tissues. (A) Metabolism of netarsudil in the presence of corneal tissue isolated from Dutch Belted rabbit (n = 3), beagle dog (n = 3), pig (n = 3), cynomolgus monkey (n = 4), and human (n = 3) corneas. Corneal metabolism assays were initiated by adding netarsudil in assay buffer to wells containing individual corneal punches in assay buffer followed by incubation at 37°C. Samples were removed at specified time intervals and analyzed by HPLC to determine the percentage of netarsudil remaining at each time point. (B) Levels of netarsudil versus netarsudil-M1 in AH following topical ocular application of netarsudil 0.02% to Dutch Belted rabbits. Netarsudil was administered to both eyes of 3 male New Zealand rabbits in each of 4 groups. In groups 1 and 2, test article was dosed only once, and samples of AH were taken from each eye by paracentesis at 4 or 6 h postdose, respectively. In groups 3 and 4, test article was dosed once daily for 3 or 4 days, respectively, and samples of AH were taken 4 h after dosing. Levels of netarsudil and its metabolite netarsudil-M1 were measured in the samples by HPLC/mass spectrometry. AH, aqueous humor; HPLC, high performance liquid chromatography.

References

    1. Alward W.L. Medical management of glaucoma. N. Engl. J. Med. 339:1298–1307, 1998
    1. Casson R.J., Chidlow G., Wood J.P.M., et al. . Definition of glaucoma: clinical and experimental concepts. Clin. Exp. Ophthalmol. 40:341–349, 2012
    1. Vrabec J.P., and Levin L.A. The neurobiology of cell death in glaucoma. Eye. 21:S11–S14, 2007
    1. Schwartz K., and Budenz D. Current management of glaucoma. Curr. Opin. Ophthalmol. 15:119–126, 2004
    1. Stamer W.D., and Acott T.S. Current understanding of conventional outflow dysfunction in glaucoma. Curr. Opin. Ophthalmol. 23:135–143, 2012
    1. Grant W.M. Clinical tonography. Trans. Am. Acad. Ophthalmol. Otolaryngol. 55:774–781, 1951
    1. Bucolo C., Platania C.B., Reibaldi M., Bonfiglio V., Longo A., Salomone S., and Drago F. Controversies in glaucoma: current medical treatment and drug development. Curr. Pharm. Des. 21:4673–4681, 2015
    1. Bucolo C., Salomone S., Drago F., Reibaldi M., Longo A., and Uva M.G. Pharmacological management of ocular hypertension: current approaches and future prospective. Curr. Opin. Pharmacol. 13:50–55, 2013
    1. Flocks M., and Zweng H.C. Studies on the mode of action of pilocarpine on aqueous outflow. Am. J. Ophthalmol. 44:380–386, 1957
    1. Barany E.H. The mode of action of pilocarpine on outflow resistance in the eye of a primate (Cercopithecus ethiops). Invest. Ophthalmol. 1:712–727, 1962
    1. Donegan R.K., and Lieberman R.L. Discovery of molecular therapeutics for glaucoma: challenges, successes, and promising directions. J. Med. Chem. 59:788–809, 2016
    1. Rao P.V., Pattabiraman P.P., and Kopczynski C. Role of the Rho GTPase/Rho kinase signaling pathway in pathogenesis and treatment of glaucoma: bench to bedside research. Exp. Eye. Res. 2016. pii:
    1. Wang S.K., and Chang R.T. An emerging treatment option for glaucoma: Rho kinase inhibitors. Clin. Ophthalmol. 8:883–890, 2014
    1. Honjo M., Tanihara H., Inatani M., et al. . Effects of rho-associated protein kinase inhibitor Y-27632 on intraocular pressure and outflow facility. Invest. Ophthalmol. Vis. Sci. 42:137–144, 2001
    1. Thieme H., Nuskovski M., Nass J.U., et al. . Mediation of calcium-independent contraction in trabecular meshwork through protein kinase C and rhoA. Invest. Ophthalmol. Vis. Sci. 41:4240–4246, 2000
    1. Rao P.V., Deng P.F., Kumar J., et al. . Modulation of aqueous humor outflow facility by the Rho kinase-specific inhibitor Y-27632. Invest. Ophthalmol. Vis. Sci. 42:1029–1037, 2011
    1. Kopczynski C.C., and Epstein D.L. Emerging trabecular outflow drugs. J. Ocul. Pharmacol. Ther. 30:85–87, 2014
    1. Garnock-Jones K.P. Ripasudil: first global approval. Drugs. 74:2211–2215, 2014
    1. Sturdivant J.M., Royalty S.M., Lin C.W., et al. . Discovery of the ROCK inhibitor netarsudil for the treatment of open-angle glaucoma. Bioorg. Med. Chem. Lett. 26:2475–2480, 2016
    1. Wang R.F., Williamson J.E., Kopczynski C., et al. . Effect of 0.04% AR-13324, a ROCK, and norepinephrine transporter inhibitor, on aqueous humor dynamics in normotensive monkey eyes. J. Glaucoma. 24:51–54, 2015
    1. Li G., Mukherjee D., Navarro I., et al. . Visualization of conventional outflow tissue responses to netarsudil in living mouse eyes. Eur. J. Pharmacol. 787:20–31, 2016
    1. Kiel J.W., and Kopczynski C. Effect of netarsudil on episcleral venous pressure in Dutch Belted rabbits. J. Ocul. Pharmacol. Ther. 31:146–151, 2015
    1. Ren R., Li G., Le T.D., et al. . Netarsudil increases outflow facility in human eyes through multiple mechanisms. Invest. Ophthalmol. Vis. Sci. 57:6197–6209, 2016
    1. Bacharach J., Dubiner H.B., Levy B., et al. ; AR-13324-CS202 Study Group. Double-masked, randomized, dose-response study of AR-13324 versus latanoprost in patients with elevated intraocular pressure. Ophthalmology. 122:302–307, 2015
    1. Lewis R.A., Levy B., Ramirez N., et al. ; PG324-CS201 Study Group. Fixed-dose combination of AR-13324 and latanoprost: a double-masked, 28-day, randomised, controlled study in patients with open-angle glaucoma or ocular hypertension. Br. J. Ophthalmol. 100:339–344, 2016
    1. Katz J.L., Weiss M.J., Heah T., et al. . Two Phase 3 studies of the efficacy and safety of AR-13324 Ophthalmic Solution 0.02%: in Patients with Open Angle Glaucoma and Ocular Hypertension. Invest. Ophthalmol. Vis. Sci. 57: 2016. [ARVO abstract]
    1. Rao P.V., and Epstein D.L. Rho GTPase/Rho kinase inhibition as a novel target for the treatment of glaucoma. BioDrugs. 21:167–177, 2007
    1. Pattabiraman P.P., Maddala R., and Rao P.V. Regulation of plasticity and fibrogenic activity of trabecular meshwork cells by Rho GTPase signaling. J. Cell. Physiol. 229:927–942, 2014
    1. Pattabiraman P.P., and Rao P.V. Mechanistic basis of Rho GTPase-induced extracellular matrix synthesis in trabecular meshwork cells. Am. J. Physiol. Cell Physiol. 298:C749–C763, 2010
    1. Draize J.H., Woodard G., and Calvery H.J. Methods for the study of irritation and toxicity of substances applied topically to the skin and mucous membranes. Pharmacol. Exp. Ther. 82:377–390, 1944
    1. Yau C.W., and Kaufman H.E. A medium-term corneal preserving medium (K-Sol). Arch. Ophthalmol. 104:598–601, 1986
    1. Tripathi R.C., Li J., Chan W.F., et al. . Aqueous humor in glaucomatous eyes contains an increased level of TGF-beta 2. Exp. Eye Res. 59:723–727, 1994
    1. Fuchshofer R. The pathogenic role of transforming growth factor-β2 in glaucomatous damage to the optic nerve head. Exp. Eye Res. 93:165–169, 2011
    1. Knipe R.S., Tager A.M., and Liao J.K. The Rho kinases: critical mediators of multiple profibrotic processes and rational targets for new therapies for pulmonary fibrosis. Pharmacol. Rev. 67:103–117, 2015
    1. Moriyama T., and Nagatoya K. The Rho-ROCK system as a new therapeutic target for preventing interstitial fibrosis. Drug News Perspect. 17:29–34, 2014
    1. Tsou P.S., Haak A.J., Khanna D., and Neubig R.R. Cellular mechanisms of tissue fibrosis. 8. Current and future drug targets in fibrosis: focus on Rho GTPase-regulated gene transcription. Am. J. Physiol. Cell. Physiol. 307:C2–C13, 2014
    1. Chang S.C., and Lee V.H. Influence of chain length on the in vitro hydrolysis of model ester prodrugs by ocular esterases. Curr. Eye Res. 2:651–656, 1982–1983
    1. Gherezghiher T., and Koss M.C. Ocular effects of dipivalyl esters of epinephrine and alpha-methylepinephrine. Exp. Eye Res. 51:537–543, 1990
    1. Rácz P., Ruzsonyi M.R., Nagy Z.T., et al. . Maintained intraocular pressure reduction with once-a-day application of a new prostaglandin F2 alpha analogue (PhXA41). An in-hospital, placebo-controlled study. Arch. Ophthalmol. 111:657–661, 1993
    1. Schroeder C., and Jordan J. Norepinephrine transporter function and human cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 303:H1273–H1282, 2012
    1. Langham M.E., and Diggs E.M. Quantitative studies of the ocular response to norepinephrine. Exp. Eye Res. 13:161–171, 1972
    1. Langham M.E. The intraocular pressure and the pupillary responses of conscious rabbits to racemic erythro-alpha-methylnorepinephrine. Exp. Eye Res. 39:781–790, 1984
    1. Pizzirani S., and Gong H. Functional anatomy of the outflow facilities. Vet. Clin. North Am. Small Anim. Pract. 45:1101–1126, 2015
    1. Levy B., Ramirez N., Novack G.D., et al. . Ocular hypotensive safety and systemic absorption of AR-13324 ophthalmic solution in normal volunteers. Am. J. Ophthalmol. 159:980–985, 2015
    1. Kopczynski C., Novack G.D., Swearingen D., et al. . Ocular hypotensive efficacy, safety and systemic absorption of AR-12286 ophthalmic solution in normal volunteers. Br. J. Ophthalmol. 97:567–572, 2013
    1. Somlyo A.P., and Somlyo A.V. Signal transduction by G-proteins, rho-kinase and protein phosphatase to smooth muscle and non-muscle myosin II. J. Physiol. 522 Pt 2:177–185, 2000

Source: PubMed

3
订阅