• 中文核心期刊要目总览
  • 中国科技核心期刊
  • 中国科学引文数据库(CSCD)
  • 中国科技论文与引文数据库(CSTPCD)
  • 中国学术期刊文摘数据库(CSAD)
  • 中国学术期刊(网络版)(CNKI)
  • 中文科技期刊数据库
  • 万方数据知识服务平台
  • 中国超星期刊域出版平台
  • 国家科技学术期刊开放平台
  • 荷兰文摘与引文数据库(SCOPUS)
  • 日本科学技术振兴机构数据库(JST)

Nitric oxide donating anti-glaucoma drugs: advances and prospects

MAO Yu-Jie, WU Jian-Bing, YANG Ze-Qiu, ZHANG Yi-Hua, HUANG Zhang-Jian

MAO Yu-Jie, WU Jian-Bing, YANG Ze-Qiu, ZHANG Yi-Hua, HUANG Zhang-Jian. Nitric oxide donating anti-glaucoma drugs: advances and prospects [J]. Chin J Nat Med, 2020, 18(4): 275-283. DOI: 10.1016/S1875-5364(20)30035-2
Citation: MAO Yu-Jie, WU Jian-Bing, YANG Ze-Qiu, ZHANG Yi-Hua, HUANG Zhang-Jian. Nitric oxide donating anti-glaucoma drugs: advances and prospects [J]. Chin J Nat Med, 2020, 18(4): 275-283. DOI: 10.1016/S1875-5364(20)30035-2

Nitric oxide donating anti-glaucoma drugs: advances and prospects

Funds: This work was supported by the National Natural Science Foundation of China (Nos. 81822041, 21977116, 81673305, and 81773573), National Science & Technology Major Project “Key New Drug Creation and Manufacturing Program” (Number: 2018ZX09711002-006-013), the open project of State Key Laboratory of Natural Medicines, No. SKLNMZZCX201824, and State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia Fund (SKL-HIDCA-2018-1), part of the work was supported by Postgraduate Research & Practice Innovation Program of Jiangsu Province (No. KYCX18_0795)
More Information
    Corresponding author:

    HUANG Zhang-Jian: E-mail: cpudahuang@163.com

  • Abstract: Glaucoma is a disease that causes irreversible blindness. Reducing intraocular pressure (IOP) is the main treatment at present. Nitric oxide (NO), an endogenous gas signaling molecule, can increase aqueous humor outflow facility, inhibit aqueous humor production thereby reducing IOP, as well as regulate eye blood flow and protect the optic nerve. Therefore, NO donating anti-glaucoma drugs have broad research prospects. In this review, we summarize NO-mediated therapy for glaucoma, and the state of the art of some NO donating molecules, including latanoprostene bunod in market and some other candidate compounds, for the intervention of glaucoma, as well as prospects and challenges ahead in this field.
  • Glaucoma is the second degenerative eye disease in the world and the leading cause of irreversible blindness. There are nearly 79.64 million glaucoma patients worldwide, and this number is estimated to reach 111.8 million by 2040 [1]. Glaucoma is characterized by optic atrophy, visual field defects and eventually progress to blindness, dividing into open-angle glaucoma and closed-angle glaucoma. Among them, primary open-angle glaucoma (POAG) is the most common one, accounting for about 74% of all glaucoma patients [2]. Increased intraocular pressure (IOP) is one of the causes of glaucoma progression. The level of IOP is controlled by the balance of aqueous humor production and outflow. Aqueous humor is produced by the ciliary epithelial cells and outflowed via two ways. One is the conventional outflow pathway, in which aqueous humor exits through the anterior ciliary vein via the trabecular meshwork (TM) and the Schlemm's canal (SC). This pathway is pressure-dependent, and elevated IOP promotes aqueous humor drainage to maintain homeostasis. The other is the unconventional outflow pathway, also known as the uveoscleral pathway, which is non-pressure-dependent. Aqueous humor enters the ciliary body and suprachoroidal space through ciliary muscle bundle space, and passes through scleral collagen or neurovascular space. At present, there are three main therapeutic methods for the intervention of glaucoma: topical medication, laser treatment and surgical intervention. Among them, topical medication is the most basic and important therapeutic mean [3]. To date, the main drugs in clinics include prostaglandin analogues, β-adrenergic receptor antagonists, α-adrenergic receptor agonists, carbonic anhydrase inhibitors and cholinergic receptor agonists [4]. Rho kinase inhibitors, as a novel class of anti-glaucoma and ocular hypertension drugs, can reduce IOP by increasing trabecular meshwork drainage and decreasing aqueous humor production, when treating alone or in combination with other anti-glaucoma drugs [5].

    Nitric oxide (NO), as a colorless, odorless gasotransmitter, and a highly active free radical molecule, plays extremely important physiological and pathological functions in cardiovascular, immune and nervous systems. NO is synthesized by three isoforms of NO synthases (NOS). Generally, under normal physiological conditions, endothelial NOS (eNOS) and neuronal NOS (nNOS) synthesize a small amount of NO (pmol·L−1−nmol·L−1) [6], while under pathological conditions, inducible NOS (iNOS) produces a large amount of NO and maintains the high concentrations of NO (μmol·L−1−mmol·L−1) for a longer time [7]. TM, SC cells, the ciliary body and iris sphincter of anterior chamber angle express nNOS and eNOS, which are associated with the regulation of blood flow and outflow of aqueous humor [7-8]. NO can promote the outflow of aqueous humor from TM and reduce IOP by activating soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway [8-9], as well as directly reduce the formation of aqueous humor via modulation of ion transporters [10]. Additionally, NO is involved in the regulation of ocular blood flow to protect visual field, and possesses neuroprotective function [11-14]. Furthermore, elevated anterior chamber perfusion pressure induces iNOS transcription to produce a large amount of NO in TM cells, which can promote the outflow of aqueous humor and downregulate IOP [15]. Notably, excessive NO may lead to pathophysiological actions such as inflammation and optic nerve degeneration via peroxynitrite and other reactive nitrogen species (RNS) which are derived from the reactions of NO with superoxide free radical or other reactive oxygen species (ROS).

    Based on the therapeutic potential of NO, NO donating compounds have attracted many attentions in the arena of glaucoma treatment. It has been reported that some typical NO donating drugs including nitroglycerin, isosorbide dinitrate, sodium nitrite, and sodium nitroprusside, can lower IOP after topical administrations [16]. Additionally, novel NO donating compounds, which could decompose to NO and the other active fragment for synergistic IOP management, have been recently developed. A representative drug Latanoprostene bunod (LBN), as a NO-donating prostaglandin F2α analogue, had been approved for the treatment of glaucoma by FDA in the USA in November 2017 [17]. Nipradilol, as a NO-donating β-adrenoceptor antagonist, was marketed in Japan in 1988 [18].

    This review addresses NO-mediated therapy for glaucoma (Fig. 1), and summarizes the state of the art of some NO donating molecules, including latanoprostene bunod in the market and some other candidate compounds, for the intervention of glaucoma, as well as prospects and challenges ahead in this field.

    Figure  1.  Summary scheme of NO-mediated therapy for glaucoma. (1) NO targets sGC to convert GTP into cGMP. cGMP activates PKG, which directly activate MLCP. Meanwhile PKG blocks the activation of RhoA to prevent inhibition of myosin phosphatase by ROCK. These actions lead to TM and SC relaxation, and increase outflow facility. (2) NO inhibits Na+ K+-ATPase in the ciliary process, and reduces aqueous humor secretion. (3) NO and downstream effectors such as cGMP participate in blood flow autoregulation. (4) NO induces S-nitrosylation of NMDA type glutamate receptors and PTEN, thus protecting RGC and axons

    As a lipophilic molecule, NO can rapidly penetrate through the phospholipid bilayer and bind to targets in cytoplasm, of which sGC is the main target [19-20]. sGC is a heterodimeric protein with heme, consisting of an α subunit and a β subunit. Binding with Fe2+ center of heme in sGC, NO activates sGC to convert guanosine triphosphate (GTP) into second messenger cGMP, which regulates the functions of many downstream effectors, such as protein kinase G (PKG), cyclic nucleotide phosphodiesterase (PDEs) and cyclic nucleotide-gated ion (CNG) channels, triggering a series of cascade reactions, such as vascular smooth muscle relaxation, platelet aggregation inhibition, myocardial contractility inhibition, and nerve excitation. In eyes, elevated PKG through NO/sGC/cGMP pathway can directly activate myosin light chain phosphatase (MLCP), which causes dephosphorylation of myosin light chain (MLC), bends protein conformation and decreases bind ability with actin, thus leading to cell relaxation [21]. In the aqueous humor outflow conventional pathway, NO induces the TM and SC cells relaxation, decrease cells volume, enlarges cells gap, decreases outflow resistance and eventually lowering IOP. Another aspect, Rho belongs to the Ras super family of low molecular weight GTPases, which consist of Rho A, Rho B, and Rho C. Rho associated coiled-coil forming protein kinase (ROCK) is one of the most typical effectors of Rho A. Activation of ROCK promotes MLC phosphorylation and increases the contractility of actin-myosin. Thus, inhibition of Rho/ROCK pathway may promote relaxation of the TM to increase aqueous outflow [22]. Sauzeau et al. found that NO can block the activation of Rho A via NO/cGMP/PKG pathway, inhibiting trabecular network contraction and promoting aqueous humor outflow [23].

    The ciliary process is a double-layer structure composed of pigmented epithelium (PE) at one side and non-pigmented epithelium (NPE) cells at the other side, which are coupled by gap junctions. PE cells connect with choroid and is responsible for the absorption of solute from blood, while NPE cells mainly secrete aqueous humor. The blood-aqueous humor barrier is located at the NPE side, preventing the backflow of aqueous humor and limiting the diffusion of proteins and other macromolecules, as well as sodium ions. Na+, K+-ATPase, as a carrier protein consisting of α and β subunits, locates on cell membrane in the ciliary process and controls the transportation of Na+ and K+. The α1 subunits, existing in the basolateral side of PE cells, play an important role in controlling the overall sodium ion secretion, while the α2 subunits, mostly concentrated on the side of NPE facing aqueous humor, control the sodium ion to enter the ciliary epithelial layer through PE. Under normal physiological conditions, Na+, K+-ATPase moves 3 sodium ions out of cell and moves 2 potassium ions into the cell to maintain the high potassium and low sodium ions liquid environment and suitable cell osmotic pressure, providing the necessary ion gradient for aqueous humor secretion and transportation. Therefore, inhibition of Na+, K+-ATPase in the ciliary process can significantly reduce the production of aqueous humor [24-25]. Early studies had demonstrated that NO and NO donor sodium nitroprusside (SNP) inhibited Na+ K+-ATPase in the choroid plexus [26], trachea [27], and kidney tubule [28]. Shahidullah et al. reported in 2001 and 2005 that SNP reduced the secretion of aqueous humor via directly acting on the ciliary epithelium (CE) cells and decreased IOP in the eyes of cattle and pigs [29-30]. In 2013, it was revealed that SNP inhibited the activity of Na+ K+-ATPase in the isolated NPE epithelial cells from pig eyes in a dose-dependent manner. Further studies found that PKG inhibitors significantly abrogated the inhibitory activity of NO donors or cGMP analogues on Na+ K+-ATPase, suggesting that the NO/sGC/cGMP/PKG pathway is involved in Na+ K+-ATPase inhibition by NO [10, 31].

    At present, there are two main mechanisms of optic nerve damage in glaucoma. One is mechanical compression, that is, optic nerve damage is caused by elevated IOP. However, at least one third of glaucoma patients possess normal IOP, suggesting that high IOP may be a symptom of glaucoma, but may not be a decisive factor. For these patients, blood flow disorders, as the other mechanism, could be particularly prominent in the process of glaucoma. Flammer et al. believed that long-term insufficiency of blood supply to the optic nerve head would lead to damage of the ganglion cells [32]. Additionally, Bata et al. observed abnormal automatic regulation of optic nerve head blood flow (ONHBF) in POAG patients. Nevertheless, there is no direct evidence that insufficient blood flow can cause glaucoma [33]. Dorner et al. found that L-NMMA, an NOS inhibitor, significantly reduced the diameter of retinal artery and vein, and decreased blood flow in choroid and retina, resulting in high IOP. These results indicated that NO can maintain the basic blood flow in eye tissue, regulate the tension of retinal blood vessels and promote the relaxation of blood vessels [11]. In addition, L-NMMA can also affect the response of ocular vascular system to the change of ocular perfusion pressure. Lasta et al. found that inhibition of NOS by L-NMMA altered the response of the retinal vein to isometric exercise, suggesting that NO participates in retinal blood flow regulation during isometric exercise [34]. Furthermore, the detailed mechanism of NO involved in ocular blood flow regulation remains to be elucidated, which may be related to the regulation of intracellular Ca2+ via by NO-sGC-cGMP pathway [35].

    As a progressive neurodegenerative disease, glaucoma causes the death of retinal ganglion cells (RGC) and axons, eventually leading to visual field defects and vision loss. The underlying mechanism is complicated with many influential factors. Direct regulation of intracellular redox state and protection of RGC from injury or death may be a new treatment for glaucoma. Mizuno et al. reported that the neuroprotective effect of nipradilol (a NO donating β receptor blocker) involved NO. This is because that NO induced S-nitrosylation of N-methyl-D-aspartate (NMDA) type glutamate receptors, thus downregulating the receptors activity and inhibiting NMDA-induced RGC damage [12]. In addition, Watanabe and Koriyama et al. found that nipradilol also induced S-nitrosylation of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which activated mammalian target of rapamycin (mTOR)/protein kinase B (Akt) pathway to promote RGC axon regeneration in cat eyes [13-14]. However, as a double-edged sword, NO could aggravate RGC death when the concentration increases. Olney et al. reported that increased IOP lead to excessive glutamate production which induced retinopathy, also known as “excitotoxicity” [36-37]. During this pathological process, over activation of NMDA receptor stimulate NOS to synthesize a large amount of NO, meanwhile, high IOP also decrease the activity of antioxidant enzymes, such as superoxide dismutase (SOD), glutathione peroxidase (GSH PX), catalase (CAT) and thioredoxin (TRX), inducing oxidative stress in RGC. The reaction of NO with superoxide free radical forms toxic peroxynitrite anion (ONOO-), accelerating RGC apoptosis via nitration mechanism [38]. Nevertheless, once glutamate concentration increases, neurons may downregulate NMDA receptors, reducing cell sensitivity to glutamate [12]. Therefore, exogenous NO release for the treatment of glaucoma should consider dosing, avoiding side effects induced by excess of NO.

    Based on the NO-mediated therapy for glaucoma, NO donating anti-glaucoma drugs have been attracted many attentions and achieved great advances. Most of NO-donating anti-glaucoma drugs consist of a conventional anti-glaucoma drug to which a NO-releasing group is attached covalently via a linker. After administration via eye drops, NO-donating anti-glaucoma drugs would decompose to regenerate parent anti-glaucoma drugs and NO, generating additional or synergistic anti-glaucoma effect. Importantly, with limited dosing, few of NO could pass blood-aqueous humor barrier to distribute whole body, avoiding side effects and enhancing druggability.

    Prostaglandins (PG), a kind of unsaturated fatty acids, play an important physiological regulatory role in blood vessels, smooth muscle, gastrointestinal tract, nervous system and respiratory system by binding with specific receptors. In eyes, among 9 subtypes of prostaglandin receptor, FP and EP1-4 are mainly distributed in the ciliary body and sclera, and less distributed in TM and SC [39]. It is found that prostaglandin F2α (PGF2α) can activate FP and EP receptors on the uveoscleral outflow pathway, relax ciliary muscle and increase aqueous outflow, thus effectively reducing IOP [40]. Latanoprost, as a PGF2α analogue, is the first anti-glaucoma drug approved by FDA in USA, which is suitable for the patients with open-angle glaucoma who are intolerant to other IOP lowering drugs. Other PGF2α analogues such as bimatoprost, travoprost and tafluprost were later approved for the treatment of glaucoma. These molecules, as ester prodrugs, activate the PG receptor in the form of free acid after hydrolysis to exert physiological activity.

    Since NO can promote aqueous outflow via the conventional pathway and prostaglandin analogues increase aqueous outflow via uveoscleral outflow pathway, NO donating prostaglandin analogs could possess two different IOP-lowering mechanisms with synergistic anti-glaucoma effect, which have attracted many attentions. Based on the structure of latanoprost, NicOx SA developed a NO donating latanoprost derivative vyzulta (latanoprostene bunod, LBN eye drops, 0.024%, Fig. 2) for the treatment of glaucoma, which was approved by FDA in 2017. After administration, LBN can be rapidly hydrolyzed in eyes to latanoprost acid (LA) and butanediol mononitrate (BDMN) (Fig. 2). LA binds to FP receptor in the eyes, upregulates the release of matrix metalloproteinase (MMP), enhances extracellular matrix digestion, and improves the outflow via uveoscleral pathway. Meanwhile, BDMN, as an NO donor moiety, is further metabolized to inactive 1, 4 butanediol and NO. It was demonstrated that LBN can increase intracellular cGMP levels by activating sGC in primary human TM cells, and LBN-mediated relaxation of TM cells was also found to be more significant than equivalent of latanoprost [41].

    Figure  2.  The structures of latanoprostene bunod, NCX 470 and the metabolic pathway

    Early studies found that transient ocular hypertensive (tOHT) rabbits were sensitive to most IOP-lowering drugs, except PGF2α receptor agonists, so the role of NO in LBN could be directly observed by using this model. In the tOHT rabbit model, LBN (0.036%) significantly decreased IOP for 13.2 mmHg during 0.5–1.5 hours after administration, while equivalent latanoprost had no effect on IOP. In both a laser-induced ocular hypertension model in nonhuman primates and a glaucoma dog model, LBN exhibited more potent IOP lowering effect than equivalent latanoprost [17]. Furthermore, in a prostaglandin FP receptor knockout mouse model, it was found that the IOP lowering effect in LBN-treated group was significantly greater than latanoprost group [42]. These experimental data in different animal models suggested that the additional IOP-lowering effect of LBN may be attributed to NO via the conventional aqueous humor outflow pathway.

    In VOYAGER phase II clinical studies, LBN (0.024%) reduced IOP more 1.23 mmHg than latanoprost (0.005%). During 28 days, LBN (0.024%) was well tolerated without aggravating ocular hyperemia, which was similar with the side effects of latanoprost (0.005%) [43-44]. In APOLLO and LUNAR Phase III studies, once-daily dosing LBN (0.024%) exhibited more potent IOP-lowering effect than twice-daily dosing timolol (0.5%) at all 9 time points over the three-month period [45-46]. In the JUPITER study, LBN was evaluated through one year of treatment among Japanese patients with OAG or OHT. The data showed that LBN treatment resulted in a significant reduction in IOP from week 4 to week 52. At week 52.9% of treated eyes had an increase in iris pigmentation [47].

    NCX 470 is another NO donating PGF2α analogue which is obtained by esterification of 15 hydroxyl in bimatoprost with 6-hydroxyhexanoic acid nitrate (Fig. 2). Similar with LBN, NCX 470 reduces IOP via uveoscleral outflow pathway mediated by bimatoprost as well as conventional aqueous humor outflow pathway triggered by NO. It was reported that NCX 470 significantly reduced IOP in tOHT rabbits, and this effect may depend on NO, not on bimatoprost and bimatoprostic acid. In both an ocular normotensive (ONT) dog model and an OHT monkey model, it was found that the IOP lowering effect of NCX 470 (0.014%) was even better than that of three equivalents of bimatoprost, suggesting the contribution of NO on IOP-lowering effect [48]. Bastia et al. reported a 28-day ocular toxicity study about NCX 470 [49]. It was showed that topical administration of NCX 470 (0.1%) to Beagle dogs in a GLP compliant condition twice daily for 28 consecutive days didn’t cause any systemic or ocular adverse findings. The results suggested adequate safety and efficacy to support advancing NCX 470 into phase 2 clinical development.

    There are two kinds of adrenergic receptors: α receptors and β receptors. β receptors are divided into β1 and β2 receptor. In eyes, α receptors are mainly distributed in the ciliary processes and β receptors are mainly distributed in the pupillary sphincter and the ciliary muscle. β-Blockers decrease the production of aqueous humor via blocking ciliary epithelial sympathetic nerve terminals, thereby reducing IOP, and the representative drugs are non-selective β-blocker timolol and β1-selective blocker betaxolol. Nipradilol, which was marketed in Japan in 1988, is a NO-donating β-adrenoceptor antagonist (Fig. 3). It was reported that the lowering IOP effect of single administration of nipradilol (0.25%) was significantly better than timolol (0.5%) in rabbit eyes. Moreover, administration of nipradilol (0.25%) twice daily for eight weeks reduced IOP by about 4 mmHg in patients with ocular hypertension, without significant change in blood pressure and heart rate. The experiment demonstrated that nipradilol had the effect of increasing uveoscleral outflow of aqueous humor and reducing aqueous humor production [50-51]. In addition, the neuroprotective effect of nipradilol is because of the production of NO, which can reduce RGC damage and promote RGC axon regeneration, as described above.

    Figure  3.  Structures of nipradilol, timolol, and NO donating timolol derivative 1

    Valerio et al. reported a novel group of NO-donating timolol derivatives which can release timolol and NO under physiologically relevant conditions. They adapted a triggering release strategy to designe new derivatives (representative compound 1) in which the enzymatic hydrolysis triggered NO donor moiety release and the subsequent intramolecular cyclization produced timolol (Fig. 3). These conjugates combining timolol- and NO-mediated properties may have better IOP lowering activity in vivo [52].

    Carbonic anhydrase (CA) mainly exists in the ciliary process, playing a major role in the regulation of aqueous humor production. Inhibition of CA reduces the secretion of bicarbonate, thus reducing the production of aqueous humor and reducing IOP [53]. Due to the distribution of CA in multiple tissues throughout the body, the first generation of oral carbonic anhydrase inhibitors (CAIs) such as acetazolamide, methazolamide, and ethoxzolamide produced systemic side effects and their clinical utilities had been progressively decreased. The second generation of topical CAIs, represented by dorzolamide and brinzolamide, became available for the treatment of ocular hypertension since 1990s (Fig. 4).

    Figure  4.  Structures of dorzolamide, brinzolamide and NO donating CAIs 2-5

    Steele et al. designed and synthesized a series of NO donating CAIs (2-3, Fig. 4) by coupling the amine group of dorzolamide with NO-donor linkers bearing nitrate ester via amide or carbamate, respectively. Interestingly, this modification did not affect the binding of compounds 2-3 with CA, producing potent CA inhibitor activity similar with dorzolamide. With NO release, compounds 2-3 possessed dual aqueous humor production inhibitory and aqueous humor outflow promoting activities. In normotensive New Zealand white rabbits, compound 2 (−4.7 mmHg) exhibited more potent IOP lowering activity than dorzolamide (−3.8 mmHg) and NO donor drug isosorbide mononitrate (ISMN, −1.9 mmHg). Meanwhile, compound 3 (120 mins) showed prolonged Tmax than dorzolamide (60 mins) [54]. Based on the cocrystal structure of dorzolamide and dorzolamide with the CA enzyme, Huang et al. selected alkyl side chains, which pointing toward the solvent, to modify nitrate ester NO donor moieties to offer compounds 4 and 5 (Fig. 4). Expectedly, 4−5 displayed good CA enzymatic potency, comparable to dorzolamide and brinzolamide. In normal rabbits, 4 and 5 exhibited improved IOP lowering activity than brinzolamide, while in OHT primate model, 5 significantly lowered IOP than brinzolamide, suggesting the potential synergistic effect of NO releasing and CA inhibition [55].

    Oxidative stress and mitochondrial dysfunction generate superoxide radicals, which react with NO to generate ONOO, thus reducing NO bioavailability. Acharya et al. designed SA-2, a NO-donating antioxidant hybrid (Fig. 5), which can protect RGCs by releasing NO as well as scavenging free radicals to exert antioxidant activity and thus increase NO bioavailability. Cell experiments showed that SA-2 upregulated SOD enzyme levels and reduced H2O2-induced cell damage in photoreceptor cells, and enhanced the cell viability against the treatment of H2O2 [56]. It was also pointed out that SA-2 spontaneously released NO in a short time at pH 7.4, which can promote angiogenesis, inhibit the proliferation and migration of smooth muscle cells and protect endothelial cells under oxidative stress [57]. SA-2 showed promising therapeutic and preventive effects in three models of acute RGC injury: hypoxia, optic nerve crush (ONC), and ischemia-reperfusion (I/R), which was mediated by upregulation of SOD enzyme levels [58].

    Figure  5.  Structures of SA-2, NCX-667, and furoxan derivatives 6 and 7

    A novel NO donor NCX 667 was reported by Bastia et al. (Fig. 5). It was effective in lowering IOP in ONT, OHT rabbit models and OHT nonhuman primate models, and multiple doses were well tolerated [59-60]. Combination of NCX 667 with the PGF2α analogue travoprost significantly reduced the IOP of normal rabbits within 5 hours after administration [61].

    Furoxan can release NO under the action of mercaptan cofactor, and the amount of NO can be regulated by the 3-substituents on the ring. The 4-substituents mainly affect the hydrophilic-lipophilic balance of the molecules. Because of the special properties of furoxan, Blangetti et al. synthesized a series of furoxan derivatives, and detected the IOP reduction effect of the target molecule in the tOHT model. It was found that compounds 6 and 7 produced similar hypotensive effects to timolol after 60 minutes of dosing, and 7 had a longer duration of action and retained activity at 120 and 240 minutes after dosing. Interestingly, after comparing the NO-releasing ability and the hydrophilic-lipophilic balance of these molecules, it was found that the antihypertensive ability of these compounds may not be determined by the amount of NO released, but depending on the hydrophilic-lipophilic balance [62].

    Besides NO donating small molecules, some researches focus on macromolecular NO delivery system to achieve sustained and prolonged NO release. Chandrawati et al. designed a liposome loaded with β-gal-NONOate, and delivered it to TM together with the carrier coated with β-galactosidase. β-gal-NOate released NO in time and dose-dependent manners through enzymatic reaction, generating IOP lowering effect [63]. In addition, based on mesoporous silica nanoparticles, Hu et al. designed a novel drug delivery system (SNP@MSNs) to deliver SNP to the TM and SC, releasing more NO over a prolonged period with IOP reduction for up to 48 hours [64]. The study of NO delivery system provides a new strategy to improve the bioavailability of NO, which also opens a new avenue for NO donating anti-glaucoma drug research.

    NO was first discovered in the 1870s, and had been ignored as a pollutant for a long time. Until 1986, Dr. Ignarro and Furchgott proposed that the endothelium-derived relaxing factor is NO, and NO became a “star molecule” and attracted more and more attentions. As outlined in Fig. 1, NO can increase aqueous humor outflow, inhibit aqueous humor generation, increase blood flow and protect RGC and axons, producing potent IOP lowering activities for the intervention of glaucoma. Compared with NO donating drugs for the treatment of other diseases, NO donating anti-glaucoma drugs possess the following advantages and features. 1) Eye drops administration could limit NO in eyes and avoid side effects mediated by wide distribution, enhancing druggability of this class of NO donating agents. As a matter of fact, LBN was the only NO donor drug approved by FDA in the last decade. Notably, naproxcinod, a NO donating nonsteroidal anti-inflammatory drug, which finished phase Ⅲ trials involving about 4 000 patients, was declined to approve by FDA in 2010, with oral administration and uncontrolled NO release as one concern. 2) It is easy to control the dose of NO released from NO donating agents by eye drops formulation, avoiding potential inflammation and optic nerve degeneration mediated by high concentrations of NO. 3) It is commonly accepted that in combination of two or three different topical anti-glaucoma drugs could realize better response of lowering IOP than monotherapy. Coupling NO donor moiety with other anti-glaucoma agents would supply more chances to achieve multiple mechanisms within one single small molecule.

    Although some advances including FDA approved drugs were achieved in this field, there are some challenges ahead in this field. Current NO donating anti-glaucoma agents are nitrate ester, so nitrate tolerance could be a potential concern for this class of anti-glaucoma agents especially for long term use. Nitrate tolerance is a phenomenon that the organic nitrates lose their hemodynamic and clinical effectiveness during sustained therapy [65]. The mechanism of nitrate tolerance remains unclear, and the main hypothesis is the gradual reduction of nitrate bioactivation which involves mitochondrial ROS formation and the mitochondrial aldehyde dehydrogenase [66]. To this end, other NO donors including furoxans, diazeniumdiolates, and metal-NO complexes warrant further studies to investigate the therapeutic potential for glaucoma. Beside classic NO/sGC/cGMP pathway, protein S-nitrosylation (SNO) could be an important mechanism for NO signaling, which could be another challenge in this field. Proteomic technologies and new SNO probes will supply more improved methods for exploring this post-translational modification to understand the mechanisms of action underlying NO-based anti-glaucoma activity [67].

    In summary, it is believed that with the deepening research on glaucoma and NO, more progress will be made in the study of NO donating anti-glaucoma agents and drugs.

  • Figure  1.   Summary scheme of NO-mediated therapy for glaucoma. (1) NO targets sGC to convert GTP into cGMP. cGMP activates PKG, which directly activate MLCP. Meanwhile PKG blocks the activation of RhoA to prevent inhibition of myosin phosphatase by ROCK. These actions lead to TM and SC relaxation, and increase outflow facility. (2) NO inhibits Na+ K+-ATPase in the ciliary process, and reduces aqueous humor secretion. (3) NO and downstream effectors such as cGMP participate in blood flow autoregulation. (4) NO induces S-nitrosylation of NMDA type glutamate receptors and PTEN, thus protecting RGC and axons

    Figure  2.   The structures of latanoprostene bunod, NCX 470 and the metabolic pathway

    Figure  3.   Structures of nipradilol, timolol, and NO donating timolol derivative 1

    Figure  4.   Structures of dorzolamide, brinzolamide and NO donating CAIs 2-5

    Figure  5.   Structures of SA-2, NCX-667, and furoxan derivatives 6 and 7

  • [1]

    Tham YC, Li X, Wong TY, et al. Global prevalence of glaucoma and projections of glaucoma burden through 2040: a systematic review and meta-analysis [J]. Ophthalmology, 2014, 121(11): 2081–2090. doi: 10.1016/j.ophtha.2014.05.013

    [2]

    Kapetanakis VV, Chan MP, Foster PJ, et al. Global variations and time trends in the prevalence of primary open angle glaucoma (POAG): a systematic review and meta-analysis [J]. Br J Ophthalmol, 2016, 100(1): 86–93. doi: 10.1136/bjophthalmol-2015-307223

    [3]

    Jonas JB, Aung T, Bourne RR, et al. Glaucoma [J]. The Lancet, 2017, 390(10108): 2183–2193. doi: 10.1016/S0140-6736(17)31469-1

    [4]

    Bucolo C, Platania CB, Reibaldi M, et al. Controversies in glaucoma: current medical treatment and drug development [J]. Curr Pharm Des, 2015, 21(32): 4673–4681. doi: 10.2174/1381612821666150909095553

    [5]

    Tanna AP, Johnson M. Rho kinase inhibitors as a novel treatment for glaucoma and ocular hypertension [J]. Ophthalmology, 2018, 125(11): 1741–1756. doi: 10.1016/j.ophtha.2018.04.040

    [6]

    Boo YC, Jo H. Flow-dependent regulation of endothelial nitric oxide synthase: role of protein kinases [J]. Am J Physiol Cell Physiol, 2003, 285(3): 499–508. doi: 10.1152/ajpcell.00122.2003

    [7]

    Becquet F, Courtois Y, Goureau O, et al. Nitric oxide in the eye: multifaceted roles and diverse outcomes [J]. Surv Ophthalmol, 1997, 42(1): 71–82. doi: 10.1016/S0039-6257(97)84043-X

    [8]

    Chang JY, Stamer WD, Bertrand J, et al. Role of nitric oxide in murine conventional outflow physiology [J]. Am J Physiol Cell Physiol, 2015, 309(4): C205–C214. doi: 10.1152/ajpcell.00347.2014

    [9]

    Behar-Cohen FF, Goureau O, D'Hermies F, et al. Decreased intraocular pressure induced by nitric oxide donors is correlated to nitrite production in the rabbit eye [J]. Invest Ophthalmol Vis Sci, 1996, 37(8): 1711–1715.

    [10]

    Shahidullah M, Mandal A, Wei G, et al. Nitric oxide regulation of Na, K-ATPase activity in ocular ciliary epithelium involves Src family kinase [J]. J Cell Physiol, 2014, 229(3): 343–352. doi: 10.1002/jcp.24454

    [11]

    Dorner GT, Garhofer G, Kiss B, et al. Nitric oxide regulates retinal vascular tone in humans [J]. Am J Physiol Heart Circ Physiol, 2003, 285(2): H631–H636. doi: 10.1152/ajpheart.00111.2003

    [12]

    Mizuno K, Koide T, Yoshimura M, et al. Neuroprotective effect and intraocular penetration of nipradilol, a beta-blocker with nitric oxide donative action [J]. Invest Ophthalmol Vis Sci, 2001, 42(3): 688–694.

    [13]

    Watanabe M, Tokita Y, Yata T. Axonal regeneration of cat retinal ganglion cells is promoted by nipradilol, an anti-glaucoma drug [J]. Neuroscience, 2006, 140(2): 517–528. doi: 10.1016/j.neuroscience.2006.02.016

    [14]

    Koriyama Y, Kamiya M, Arai K, et al. Nipradilol promotes axon regeneration through S-nitrosylation of PTEN in retinal ganglion cells [J]. Adv Exp Med Biol, 2014, 801: 751–757.

    [15]

    Schneemann A, Leusink-Muis A, van den Berg T, et al. Elevation of nitric oxide production in human trabecular meshwork by increased pressure [J]. Graefes Arch Clin Exp Ophthalmol, 2003, 241(4): 321–326. doi: 10.1007/s00417-003-0638-4

    [16]

    Nathanson JA. Nitrovasodilators as a new class of ocular hypotensive agents [J]. J Pharmacol Exp Ther, 1992, 260(3): 956–965.

    [17]

    Krauss AH, Impagnatiello F, Toris CB, et al. Ocular hypotensive activity of BOL-303259-X, a nitric oxide donating prostaglandin F2α agonist, in preclinical models [J]. Exp Eye Res, 2011, 93(3): 250–255. doi: 10.1016/j.exer.2011.03.001

    [18]

    Taguchi R, Shirakawa H, Yamaguchi T, et al. Nitric oxide-mediated effect of nipradilol, an α-and β-adrenergic blocker, on glutamate neurotoxicity in rat cortical cultures [J]. Eur J Pharmacol, 2006, 535(1−3): 86–94. doi: 10.1016/j.ejphar.2006.01.036

    [19]

    Knowles RG, Palacios M, Palmer RM, et al. Formation of nitric oxide from L-arginine in the central nervous system: a transduction mechanism for stimulation of the soluble guanylate cyclase [J]. Proc Natl Acad Sci USA, 1989, 86(13): 5159–5162. doi: 10.1073/pnas.86.13.5159

    [20]

    Mergia E, Friebe A, Dangel O. Spare guanylyl cyclase NO receptors ensure high NO sensitivity in the vascular system [J]. J Clin Invest, 2006, 116(6): 1731–1737.

    [21]

    Gao N, Tsai MH, Chang AN, et al. Physiological vs pharmacological signalling to myosin phosphorylation in airway smooth muscle [J]. J Physiol, 2017, 595(19): 6231–6247. doi: 10.1113/JP274715

    [22]

    Wettschureck N, Offermanns S. Rho/Rho-kinase mediated signaling in physiology and pathophysiology [J]. J Mol Med-JMM, 2002, 80(10): 629–638.

    [23]

    Sauzeau V, Le Jeune H, Cario-Toumaniantz C, et al. Cyclic GMP-dependent protein kinase signaling pathway inhibits RhoA-induced Ca2+ sensitization of contraction in vascular smooth muscle [J]. J Biol Chem, 2000, 275(28): 21722–21729. doi: 10.1074/jbc.M000753200

    [24]

    Arakaki X, McCleary P, Techy M, et al. Na, K-ATPase alpha isoforms at the blood-cerebrospinal fluid-trigeminal nerve and blood-retina interfaces in the rat [J]. Fluids Barriers CNS, 2013, 10(1): 14.

    [25]

    Ghosh S, Hernando N, Martín-Alonso JM, et al. Expression of multiple Na+, K+-ATPase genes reveals a gradient of isoforms along the nonpigmented ciliary epithelium: functional implications in aqueous humor secretion [J]. J Cell Physiol, 1991, 149(2): 184–194. doi: 10.1002/jcp.1041490203

    [26]

    Ellis DZ, Nathanson JA, Sweadner KJ. Carbachol inhibits Na+, K+-ATPase activity in choroid plexus via stimulation of the NO/cGMP pathway [J]. Am J Physiol-cell Ph, 2000, 279(6): C1685–C1693.

    [27]

    Elias MD, de Lima WT, Vannuchi YB, et al. Nitric oxide modulates Na+, K+-ATPase activity through cyclic GMP pathway in proximal rat trachea [J]. Eur J Pharmacol, 1999, 367(2−3): 307–314.

    [28]

    Seven I, Turkozkan N, Cimen B. The effects of nitric oxide synthesis on the Na+, K+-ATPase activity in guinea pig kidney exposed to lipopolysaccharides [J]. Mol Cell Biochem, 2005, 271(1−2): 107–112.

    [29]

    Millar JC, Shahidullah M, Wilson WS. Intraocular pressure and vascular effects of sodium azide in bovine perfused eye [J]. J Ocul Pharmacol Ther, 2001, 17(3): 225–234.

    [30]

    Shahidullah M, Yap M, To CH. Cyclic GMP, sodium nitroprusside and sodium azide reduce aqueous humour formation in the isolated arterially perfused pig eye [J]. Br J Pharmacol, 2005, 145(1): 84–92.

    [31]

    Shahidullah M, Delamere NA. NO donors inhibit Na, K-ATPase activity by a protein kinase G-dependent mechanism in the nonpigmented ciliary epithelium of the porcine eye [J]. Br J Pharmacol, 2006, 148(6): 871–880.

    [32]

    Flammer J. The vascular concept of glaucoma [J]. Surv Ophthalmol, 1994, 38 Suppl: S3–S6.

    [33]

    Bata AM, Fondi K, Witkowska KJ, et al. Optic nerve head blood flow regulation during changes in arterial blood pressure in patients with primary open-angle glaucoma [J]. Acta Ophthalmol, 2019, 97(1): e36–e41. doi: 10.1111/aos.13850

    [34]

    Lasta M, Polak K, Luksch A, et al. Effect of NO synthase inhibition on retinal vessel reaction to isometric exercise in healthy humans [J]. Acta Ophthalmol, 2012, 90(4): 362–368. doi: 10.1111/j.1755-3768.2010.01970.x

    [35]

    Carvajal JA, Germain AM, Huidobro-Toro JP, et al. Molecular mechanism of cGMP-mediated smooth muscle relaxation [J]. J Cell Physiol, 2000, 184(3): 409–420. doi: 10.1002/1097-4652(200009)184:3<409::AID-JCP16>3.0.CO;2-K

    [36]

    Olney JW. Glutaate-induced retinal degeneration in neonatal mice. Electron microscopy of the acutelyevolving lesion [J]. J Neuropathol Exp Neurol, 1969, 28(3): 455–474. doi: 10.1097/00005072-196907000-00007

    [37]

    Olney JW, Ho OL. Brain damage in infant mice following oral intake of glutamate, aspartate or cysteine [J]. Nature, 1970, 227(5258): 609–611.

    [38]

    Nita M, Grzybowski A. The role of the reactive oxygen species and oxidative stress in the pathomechanism of the age-related ocular diseases and other pathologies of the anterior and posterior eye segments in adults [J]. Oxid Med Cell Longev, 2016, 2016: 3164734.

    [39]

    Alexander SP, Christopoulos A, Davenport AP, et al. The concise guide to pharmacology 2017/18: G protein-coupled receptors [J]. Br J Pharmacol, 2017, 174 (S1): S17–S129.

    [40]

    Winkler NS, Fautsch MP. Effects of prostaglandin analogues on aqueous humor outflow pathways [J]. J Ocul Pharmacol Ther, 2014, 30(2−3): 102–109. doi: 10.1089/jop.2013.0179

    [41]

    Cavet ME, Vollmer TR, Harrington KL, et al. Regulation of endothelin-1-induced trabecular meshwork cell contractility by latanoprostene bunod [J]. Invest Ophthalmol Vis Sci, 2015, 56(6): 4108–4116. doi: 10.1167/iovs.14-16015

    [42]

    Saeki T, Tsuruga H, Aihara M, et al. Dose-response profile of PF-03187207(PF-207) and peak IOP lowering response following single topical administration to FP receptor knockout mice vs wild type mice [J]. ARVO Annual Meeting Abstract, 2009, 50: 4064.

    [43]

    Weinreb RN, Ong T, Sforzolini BS, et al. A randomised, controlled comparison of latanoprostene bunod and latanoprost 0.005% in the treatment of ocular hypertension and open angle glaucoma: The VOYAGER study [J]. Br J Ophthalmol, 2015, 99(6): 738–745. doi: 10.1136/bjophthalmol-2014-305908

    [44]

    Govoni M, Casagrande S, Maucci R, et al. In vitro metabolism of (nitrooxy) butyl ester nitric oxide-releasing compounds: comparison with glyceryl trinitrate [J]. J Pharmacol Exp Ther, 2006, 317(2): 752–761. doi: 10.1124/jpet.105.097469

    [45]

    Weinreb RN, Scassellati Sforzolini B, Vittitow J, et al. Latanoprostene bunod 0.024% versus timolol maleate 0.5% in subjects with open-angle glaucoma or ocular hypertension: the APOLLO study [J]. Ophthalmology, 2016, 123(5): 965–973. doi: 10.1016/j.ophtha.2016.01.019

    [46]

    Medeiros FA, Martin KR, Peace J, et al. Comparison of latanoprostene bunod 0.024% and timolol maleate 0.5% in open-angle glaucoma or ocular hypertension: the LUNAR study [J]. Am J Ophthalmol, 2016, 168: 250–259. doi: 10.1016/j.ajo.2016.05.012

    [47]

    Kawase K, Vittitow JL, Weinreb RN, et al. Long-term safety and efficacy of latanoprostene bunod 0.024% in Japanese subjects with open-angle glaucoma or ocular hypertension: the JUPITER study [J]. Adv Ther, 2016, 33(9): 1612–1627. doi: 10.1007/s12325-016-0385-7

    [48]

    Impagnatiello F, Toris CB, Batugo M, et al. Intraocular pressure-lowering activity of NCX 470, a novel nitric oxide-donating bimatoprost in preclinical models [J]. Invest Ophthalmol Vis Sci, 2015, 56(11): 6558–6564. doi: 10.1167/iovs.15-17190

    [49]

    Bastia E, Almirante N, Bergamini MVW, et al. Nonclinical development of NCX 470, a novel nitric oxide (NO)-donating, IOP lowering prostaglandin analog for glaucoma and ocular hypertension [J]. ARVO Annual Meeting Abstract, 2019, 60: 3806.

    [50]

    Kanno M, Araie M, Tomita K, et al. Effects of topical nipradilol, a beta-blocking agent with alpha-blocking and nitroglycerin-like activities, on aqueous humor dynamics and fundus circulation [J]. Invest Ophthalmol Vis Sci, 1998, 39(5): 736–743.

    [51]

    Kanno M, Araie M, Koibuchi H, et al. Effects of topical nipradilol, a beta blocking agent with alpha blocking and nitroglycerin-like activities, on intraocular pressure and aqueous dynamics in humans [J]. Br J Ophthalmol, 2000, 84(3): 293–299. doi: 10.1136/bjo.84.3.293

    [52]

    Valerio C, Minerva RB, Stefano B, el al. Synthesis of novel nitric oxide (NO)-releasing esters of timolol [J]. Bioorg Med Chem Lett, 2009, 19(10): 2785–2788. doi: 10.1016/j.bmcl.2009.03.111

    [53]

    Mincione F, Scozzafava A, Supuran CT. The development of topically acting carbonic anhydrase inhibitors as anti-glaucoma agents [J]. Curr Pharm Des, 2008, 14(7): 649–654. doi: 10.2174/138161208783877866

    [54]

    Steele RM, Benedini F, Biondi S, et al. Nitric oxide-donating carbonic anhydrase inhibitors for the treatment of open-angle glaucoma [J]. Bioorg Med Chem Lett, 2009, 19(23): 6565–6570. doi: 10.1016/j.bmcl.2009.10.036

    [55]

    Huang Q, Rui EY, Cobbs M, et al. Design, synthesis, and evaluation of NO-donor containing carbonic anhydrase inhibitors to lower intraocular pressure [J]. J Med Chem, 2015, 58(6): 2821–2833. doi: 10.1021/acs.jmedchem.5b00043

    [56]

    Acharya S, Rogers P, Krishnamoorthy RR, et al. Design and synthesis of novel hybrid sydnonimine and prodrug useful for glaucomatous optic neuropathy [J]. Bioorg Med Chem Lett, 2016, 26(5): 1490–1494. doi: 10.1016/j.bmcl.2015.12.030

    [57]

    Le DQ, Kuriakose AE, Nguyen DX, et al. Hybrid nitric oxide donor and its carrier for the treatment of peripheral arterial diseases [J]. Sci Rep, 2017, 7: 8692. doi: 10.1038/s41598-017-08441-9

    [58]

    Stankowska DL, Dibas A, Li L, et al. Hybrid compound SA-2 is neuroprotective in animal models of retinal ganglion cell death [J]. Invest Ophthalmol Vis Sci, 2019, 60(8): 3064–3073. doi: 10.1167/iovs.18-25999

    [59]

    Bastia E, Impagnatiello F, Almirante N, et al. NCX 667, a novel nitric oxide (NO) donor lowers intraocular pressure (IOP) in ocular normotensive and hypertensive eyes of rabbits and nonhuman primates [J]. ARVO Annual Meeting Abstract, 2015, 59: 1999.

    [60]

    Bastia E, Impagnatiello F, Ongini E, et al. Repeated dosing of NCX 667, a new nitric oxide (NO) donor, retains IOP-lowering activity in animal models of glaucoma [J]. ARVO Annual Meeting Abstract, 2017, 58: 2106.

    [61]

    Bastia E, Masini E, Durante M, et al. IOP-lowering effects of NCX 667 in combination with travoprost in ocular normotensive and transient hypertensive rabbits [J]. ARVO Annual Meeting Abstract, 2016, 57: 3031.

    [62]

    Blangetti M, Rolando B, Chegaev K, et al. New furoxan derivatives for the treatment of ocular hypertension [J]. Bioorg Med Chem Lett, 2017, 27(3): 479–483. doi: 10.1016/j.bmcl.2016.12.041

    [63]

    Chandrawati R, Chang JYH, Reina-Torres E, et al. Localized and controlled delivery of nitric oxide to the conventional outflow pathway via enzyme biocatalysis: toward therapy for glaucoma [J]. Adv Mater, 2017, 29(16): 1604932. doi: 10.1002/adma.201604932

    [64]

    Hu CC, Sun JG, Zhang Y, et al. Local delivery and sustained-release of nitric oxide donor loaded in mesoporous silica particles for efficient treatment of primary open-angle glaucoma [J]. Adv Healthc Mater, 2018, 7(23): 1801047. doi: 10.1002/adhm.201801047

    [65]

    Napoli C, Ignarro LJ. Nitric oxide and atherosclerosis [J]. Nitric Oxide, 2001, 5(2): 88–97. doi: 10.1006/niox.2001.0337

    [66]

    Daiber A, Wenzel P, Oelze M, et al. New insights into bioactivation of organic nitrates, nitrate tolerance and cross-tolerance [J]. Clin Res Cardiol, 2008, 97(1): 12–20. doi: 10.1007/s00392-007-0588-7

    [67]

    López-Sánchez LM, López-Pedrera C, Rodríguez-Ariza A. Proteomic approaches to evaluate protein s-nitrosylation in disease [J]. Mass Spectrom Rev, 2014, 33(1): 7–20. doi: 10.1002/mas.21373

  • 期刊类型引用(28)

    1. Liu, J., Liang, Y., Tian, Y. et al. Unlocking the Untapped Potential of Photoresponsive Dual-Gas Nanomedicine in Glaucoma Therapy. Advanced Healthcare Materials, 2025, 14(12): 2404245. 必应学术
    2. Liu, J., Lu, Y., Tian, Y. et al. A Light-Activatable Nitric Oxide Donor for Targeted Glaucoma Therapy with Real-Time Monitoring Capabilities. Advanced Healthcare Materials, 2025, 14(11): 2404221. 必应学术
    3. Li, Z., Qin, F., Liu, C. et al. Alleviating heat stress-induced immune organ damage in ducks: Role of melittin. Tropical Animal Health and Production, 2025, 57(2): 57. 必应学术
    4. Adebayo, A., Adebayo, E. Not a laughing matter: revisiting nitric oxide as a potential target for glaucoma therapy. Medical Gas Research, 2025, 15(1): 112–113. 必应学术
    5. Li, C., Zhu, M., Liu, S. et al. Development of Nitric Oxide-Donating Netarsudil Derivatives as a Synergistic Therapy for Glaucoma with Reduced Ocular Irritation. Journal of Medicinal Chemistry, 2024, 67(18): 16311–16327. 必应学术
    6. Nakamura, R., Ciranna, N.S., Fernández, J.C. et al. Methylene Blue Reduces Electroretinogram Distortion and Ganglion Cell Death in a Rat Model of Glaucoma. Biomedicines, 2024, 12(9): 1983. 必应学术
    7. Park, H.S., Kim, J.W. Effect of Latanoprostene Bunod on the Permeability of Trabecular Meshwork Cells. Journal of Korean Ophthalmological Society, 2024, 65(2): 139–144. 必应学术
    8. Benitez, B., Anter, A.M., Arcuri, J. et al. Currently available prostanoids for the treatment of glaucoma and ocular hypertension: A review. Current Opinion in Pharmacology, 2024, 74: 102424. 必应学术
    9. Wang, Y., Sun, M., Li, Y. et al. Hydrostatic Pressure Induces Activation of Astrocytes through the JAK2/STAT3 Pathway. Journal of Biological Regulators and Homeostatic Agents, 2024, 38(3): 2435–2446. 必应学术
    10. Zhou, X., Zhang, F., Zhang, X. et al. Construction of miRNA-mRNA regulatory network indicates potential biomarkers for primary open-angle glaucoma. BMC Medical Genomics, 2023, 16(1): 280. 必应学术
    11. Rizvi, A.A., Abbas, M., Ansari, A.I. et al. Impact of Type 2 Diabetes Mellitus-Related Factors on Glaucoma: a Comprehensive Review. Current Ophthalmology Reports, 2023, 11(4): 69–77. 必应学术
    12. Wang, X., Wang, M., Liu, H. et al. The Association between Vascular Abnormalities and Glaucoma—What Comes First?. International Journal of Molecular Sciences, 2023, 24(17): 13211. 必应学术
    13. Naumenko, L.V., Taran, A.S., Shevchenko, A.A. PROSPECTS FOR DRUG THERAPY OF GLAUCOMA | ÏÅÐÑÏÅÊÒÈÂÛ ËÅÊÀÐÑÒÂÅÍÍÎÉ ÒÅÐÀÏÈÈ ÃËÀÓÊÎÌÛ. Eksperimental'naya i Klinicheskaya Farmakologiya, 2023, 86(11): 34–39. 必应学术
    14. Ruiz-Lozano, R.E., Azar, N.S., Mousa, H.M. et al. Ocular surface disease: a known yet overlooked side effect of topical glaucoma therapy. Frontiers in Toxicology, 2023, 5: 1067942. 必应学术
    15. Soundararajan, L., Dharmarajan, A., Samji, P. Regulation of pleiotropic physiological roles of nitric oxide signaling. Cellular Signalling, 2023, 101: 110496. 必应学术
    16. Huang, Y.-L., Hsu, Y.-L., Yu, Y.-C. et al. A systematic approach to reduce intraocular pressure for the treatment of glaucoma. Biotechnology Progress, 2022, 38(6): e3285. 必应学术
    17. Yang, Z., Wu, J., Wu, K. et al. Identification of Nitric Oxide-Donating Ripasudil Derivatives with Intraocular Pressure Lowering and Retinal Ganglion Cell Protection Activities. Journal of Medicinal Chemistry, 2022, 65(17): 11745–11758. 必应学术
    18. Lo, T.-C., Chen, Y.-Y., Hung, M.-C. et al. Latanoprostene Bunod 0.024% in the Treatment of Open-Angle Glaucoma and Ocular Hypertension: A Meta-Analysis. Journal of Clinical Medicine, 2022, 11(15): 4325. 必应学术
    19. Li, Y., Lu, H., Sun, Y. Correlation of NO and ET-1 Levels with Blood Pressure Changes in Hemodialysis Patients after Arteriovenous Fistula Surgery. Frontiers in Surgery, 2022, 9: 905372. 必应学术
    20. Kulkarni, A., Kamath, Y., Shetty, L. et al. The Effect of Specific Techniques of Nasal Breathing [Pranayama] on Intra-Ocular Pressure in Normal Individuals, a Randomized Trial. Clinical Ophthalmology, 2022, 16: 4047–4054. 必应学术
    21. Zhou, J., Zeng, W., Zeng, Y. et al. Anticancer and Anti-Inflammatory Mechanisms of NOSH-Aspirin and Its Biological Effects. Computational and Mathematical Methods in Medicine, 2022. 必应学术
    22. Wu, X., Yang, X., Liang, Q. et al. Drugs for the treatment of glaucoma: Targets, structure-activity relationships and clinical research. European Journal of Medicinal Chemistry, 2021, 226: 113842. 必应学术
    23. Wang, T., Cao, L., Jiang, Q. et al. Topical Medication Therapy for Glaucoma and Ocular Hypertension. Frontiers in Pharmacology, 2021, 12: 749858. 必应学术
    24. Han, B., Song, M., Li, L. et al. The application of nitric oxide for ocular hypertension treatment. Molecules, 2021, 26(23): 7306. 必应学术
    25. Luo, Y., Zhu, Y., Basang, W. et al. Roles of Nitric Oxide in the Regulation of Reproduction: A Review. Frontiers in Endocrinology, 2021, 12: 752410. 必应学术
    26. Storgaard, L., Tran, T.L., Freiberg, J.C. et al. Glaucoma Clinical Research: Trends in Treatment Strategies and Drug Development. Frontiers in Medicine, 2021, 8: 733080. 必应学术
    27. Sayed, S., Abdel-Moteleb, M., Amin, M.M. et al. Cubogel as potential platform for glaucoma management. Drug Delivery, 2021, 28(1): 293–305. 必应学术
    28. HUANG, Z.-J., WANG, B.-H. Gaseous signaling molecules as potential therapeutic agents. Chinese Journal of Natural Medicines, 2020, 18(4): 273–274. 必应学术

    其他类型引用(0)

图(5)
计量
  • 文章访问数:  5778
  • HTML全文浏览量:  1517
  • PDF下载量:  59
  • 被引次数: 28
出版历程
  • 收稿日期:  2020-01-04
  • 网络出版日期:  2020-03-23
  • 刊出日期:  2020-03-31

目录

/

返回文章
返回