Next Article in Journal
Prevalence of Genetic Diamine Oxidase (DAO) Deficiency in Female Patients with Fibromyalgia in Spain
Previous Article in Journal
Breaking a Vicious Circle: Lymphangiogenesis as a New Therapeutic Target in Wound Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Therapies for the Prevention of Fibrosis in Glaucoma Filtration Surgery

1
School of Medicine, University of Missouri, Columbia, MO 65212, USA
2
Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA
3
One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
4
Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(3), 657; https://doi.org/10.3390/biomedicines11030657
Submission received: 28 December 2022 / Revised: 7 February 2023 / Accepted: 15 February 2023 / Published: 21 February 2023
(This article belongs to the Section Molecular and Translational Medicine)

Abstract

:
Conjunctival fibrosis remains the major impediment to the success of glaucoma filtration surgery. Anti-metabolites remain the gold standard for mitigating post-surgical fibrosis, but they are associated with high complication rates and surgical failure rates. Establishing a more targeted approach to attenuate conjunctival fibrosis may revolutionize the surgical approach to glaucoma. A new strategy is needed to prevent progressive tissue remodeling and formation of a fibrotic scar, subsequently increasing surgical success and reducing the prevalence of glaucoma-related vision loss. Advancements in our understanding of molecular signaling and biomechanical cues in the conjunctival tissue architecture are broadening the horizon for new therapies and biomaterials for the mitigation of fibrosis. This review aims to highlight the strategies and current state of promising future approaches for targeting fibrosis in glaucoma filtration surgery.

Graphical Abstract

1. Introduction

Glaucoma is a progressive, degenerative disease of the optic nerve that can result in severe visual field loss and blindness. It is the leading cause of irreversible blindness worldwide [1,2,3,4,5,6]. The overall worldwide prevalence of primary open-angle glaucoma, the most common form of glaucoma, is 2.4% (95% CI 2.0~2.8%), and it affects an estimated 68 million people worldwide [7].
Currently, the only modifiable risk factor and proven therapy to prevent the progression of glaucoma is lowering the intraocular pressure (IOP). Reduction of IOP can be accomplished with topical eye drops, oral medications, or laser procedures that increase aqueous outflow or reduce aqueous production. The surgical treatment of glaucoma similarly decreases aqueous inflow or increases aqueous outflow. The latter can be accomplished by augmenting existing outflow pathways or creating artificial outflow pathways into the subconjunctival or sub-Tenon’s space, often referred to as traditional incisional glaucoma filtration surgery. Micro-invasive glaucoma surgical (MIGS) approaches allow for enhancement of the conventional aqueous outflow pathways through the trabecular meshwork and into Schlemm’s canal or alternatively from the anterior chamber into the suprachoroidal space. Traditional incisional glaucoma filtering surgery allows the aqueous humor to drain into the space between the sclera and conjunctiva and/or Tenon’s tissues.
Unlike many surgeries, where the goal is to heal tissue with the restoration of normal architecture, traditional incisional glaucoma filtering surgery aims to modulate wound healing. Wound healing modulation allows for continued aqueous egress into the subconjunctival and/or sub-Tenon’s space, thereby preventing surgical failure [8,9]. Episcleral and subconjunctival fibrosis remain major impediments to the success of glaucoma filtering surgery. Conjunctival fibrosis at the site of filtration may impede aqueous outflow, leading to inadequate IOP reduction. Surgical failure is associated with poor postoperative IOP control, consequent progression of glaucomatous disc cupping, and visual field loss [10]. 5-fluorouracil (5-FU) and mitomycin-C (MMC) are widely used to attenuate ocular fibrosis. The usage of 5-FU and MMC have led to improved surgical success rates, but with a concomitant increase in postoperative complications. Complications associated with broad non-specific antifibrotics include wound leaks, hypotony, and infection [11]. Therefore, broadening the therapeutic approach for glaucoma filtration surgery is necessary, and agents with more specific physiological actions and less cytotoxicity are needed. This review aims to highlight the current understanding of potential therapeutic targets for the prevention of fibrosis and discuss novel methods to improve glaucoma surgical success.

2. Causes of Ocular Fibrosis following Glaucoma Filtration Surgery

2.1. Wound Healing

Like other surgeries, glaucoma filtration surgery induces tissue trauma. Crosstalk among various players such as growth factors, cytokines, signaling pathways, and immune regulators is critical during conjunctival wound healing. Wound healing has been defined in three distinct and overlapping phases: the inflammatory, proliferative, and maturation stages (Figure 1) [10,12,13,14]. In the inflammatory phase, plasma proteins and blood cells migrate to the site of injury. During this phase, damaged cells and pathogens are removed while bleeding is controlled. The resultant clot, platelet plug hormones, cytokines, and growth factors attract neutrophils, macrophages, and lymphocytes to the site of injury. IL-1, Interferon-α2b, and growth factors such as transforming growth factor β (TGFβ), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) are among these mediators [10].
In the proliferative phase, endothelial cells and fibroblasts migrate to the site of injury. The key processes in this phase are angiogenesis and granulation tissue formation. Fibroblasts increase in number, while neutrophils and monocytes secrete proteolytic enzymes and promote debridement. Activated phagocytes increase growth factors and cytokines such as TGFβ to activate and maintain fibroblasts. VEGF promotes blood vessel formation, and PDGF stimulates fibroblasts [12,13].
In the maturation phase, scar tissue is formed during tissue remodeling as blood vessels regress over time and fibroblasts induce crosslinking of collagen type I and elastin. This phase is defined by matrix metalloproteinases (MMPs), which are synthesized by macrophages, neutrophils, and fibroblasts. This synthesis leads to collagen supercoils and the transformation of granulation tissue into scar tissue [10]. The reduction in myofibroblast number is also key to this phase, as their prolonged survival leads to excessive scarring [13]. Modulation of scarring may target many points of fibrosis regulation, including the migration, proliferation, myofibroblast transdifferentiation, and apoptosis of fibroblasts [15].

2.2. Growth Factors and Cytokines

TGFβ, PDGF, and VEGF are cytokines that play a key role in modulating ocular fibrosis [16]. Tissue damage transforms TGFβ into its active form. Fibroblasts migrate to the surgical site and are transformed into extracellular matrix (ECM)-producing myofibroblasts, the precursors for Tenon’s fibroblasts, the most important mediator of the development of conjunctival fibrosis under the scleral flap. TGFβ is a potent inducer of Tenon’s fibroblast proliferation, migration, and collagen production. TGFβ also stimulates angiogenesis and acts as a chemoattractant to other macrophages and fibroblasts [17]. PDGF induces the proliferation of fibroblasts and migration to the site of injury [18], and it is upregulated in fibrosis. VEGF plays a key role in promoting cell proliferation. It is a potent mediator of angiogenesis, vasculogenesis, and vascular endothelial cell permeability, all of which are critical processes in wound healing. The signaling pathways are discussed in the next section.

2.3. Intracellular Signaling Driving Fibrosis

2.3.1. TGFβ

Signal transduction through the effects of TGFβ has been extensively studied. TGFβ has been targeted for its inflammatory and proliferative effects. TGFβ is found in high concentrations in the aqueous humor of glaucoma patients [19,20] and in the trabecular meshwork [21]. Thus, it is likely that it has a role in the regulation of aqueous outflow. Several studies have shown that TGFβ activates fibroblast proliferation in vitro and in vivo [14,22,23,24]. Roberts et al. demonstrated the proliferative effects of TGFβ by injecting TGFβ into murine models, which resulted in fibroblast proliferation and collagen deposition [23].
TGFβ2 is the predominant isoform found in the eye and accounts for the greatest stimulation of fibrosis. Due to the robust amount of TGFβ in fibroblasts, targeting this pathway holds promise for improving outcomes of glaucoma surgery. However, blocking all TGFβ isoforms is excessive, as some TGFβ is essential for normal wound healing. Alternatively, the use of a monoclonal antibody directed solely against TGFβ2 may be too narrow of an approach due to the involvement of other TGFβ isoforms in ocular fibrosis.
TGFβ exerts its effects through serine/threonine and tyrosine kinase receptors. TGFβ signals through small mothers against decapentaplegic (SMAD)-independent and SMAD-dependent pathways [25]. Crosstalk between these pathways has been reported. In SMAD-dependent signaling, SMAD proteins transduce signals from the membrane to the nucleus. SMAD proteins are known to mediate the transcription of various genes for the transdifferentiation of fibroblasts to myofibroblasts [26].
SMAD 2, 3, and 4 proteins are specific for TGFβ signaling [27,28]. SMAD 2 and 3 are phosphorylated by the TGFβ type I receptor kinase and partner with SMAD 4. This complex is translocated to the nucleus, where transcription products are targeted. SMAD 3 is crucial to wound healing in many tissues. SMAD 7 is inhibitory. It is induced by TGFβ as well as other cytokines, and it blocks SMAD 2 and SMAD 3 signaling [29].
In SMAD-independent signaling, JNK, p38, mTOR, and ROCK pathways are responsible for apoptosis and actin cytoskeleton reorganization. P38 MAPK is also critical for cell migration. JNK also mediates connective tissue growth factor (CTGF) expression [30].

2.3.2. PDGF

PDGF is another cytokine implicated in the progression of wound healing. PDGF is a dimer that can exist as a homo- or heterodimer made up of A and B subunits. PDGF stimulates keratocyte proliferation, survival, and migration. Thus, PDGF upregulation has been associated with pathological angiogenesis and fibrotic conditions. PDGF acts synergistically with TGFβ in an autocrine manner to stimulate differentiation of the myofibroblast [31].

2.3.3. FGF2

FGF2 also promotes the proliferation and differentiation of keratocytes into fibroblasts [31] and is a possible therapeutic target to inhibit fibrosis. Unlike PDGF, FGF2 downregulates alpha-smooth muscle actin (α-SMA), which allows for wound contraction, in myofibroblasts [32].

2.4. Biomechanical Cues

In addition to biochemical modulation, biomechanical cues play a significant role in scar formation. Increased tissue stiffness facilitates myofibroblast transdifferentiation and fibrosis [14]. Morphologically, myofibroblasts have a contractile apparatus that contains bundles of actin microfilaments with contractile proteins. A distinguishing feature of the myofibroblast is the expression of α-SMA. The α-SMA adds to the mechanical strength of the cell and allows for wound contraction. This provides a mechano-transduction system, as the force that is generated by the fibers can be transmitted to the ECM. In addition, extracellular mechanical signals can be transduced into intracellular signals through this mechano-transduction system. Topographic features and substrate compliance are all involved in the modulation of myofibroblast differentiation. Increased expression of α-SMA is correlated with increased force generation by myofibroblasts. It has subsequently been shown that most myofibroblasts express α-SMA, and the expression of α-SMA and collagen type I in these cells is coordinated by TGFβ1. Thus, the myofibroblast plays a key role in the synthesis of ECM and force generation, which results in ECM reorganization and wound contraction [33].
It is widely accepted that the cellular response is governed by the distribution of mechanical forces throughout the tissue. Although the mechanosensitive cellular components are important to the mechanical responses in fibrosis, understanding the context of the response to glaucoma surgery may help to target new therapies in addressing the high failure and complication rates in this surgery [34].
Coordinating the response from physical force to biochemical information is essential to tissue healing [35]. Force can change the response to glaucoma filtration surgery in that wall stress is proportional to capsule thickness in glaucoma surgery [36]. Increased tissue stiffness facilitates myofibroblast transdifferentiation and subsequent fibrosis. Similarly, after phototherapeutic keratectomy, elevated stiffness of the anterior corneal stroma precedes the upregulation of myofibroblasts, and this stiffness does not return to pre-surgical values [37]. In vitro, stromal cells treated with TGFβ1 were stiffer than untreated cells. The stiffness of these cells has been shown to depend on the substrate stiffness. Thus, the geometry of biomaterials and implantable devices impacts the extent of fibrosis, and the stiffness of the microenvironment surrounding the incision can change cellular mechanics [36,37]. The remodeling of ECM fibers by fibroblasts in interstitial flow also contributes to the extent of fibrosis, as a perpendicular alignment can also decrease shear and drag forces, decrease tissue permeability, and increase hydrostatic pressure [38]. The filtering bleb after filtration surgery is subjected to mechanical stress from the draining of aqueous humor. Further investigation into the integration between biomechanical and biochemical signaling is warranted. These studies will allow for the development of new therapies that consider the biochemical and biophysical microenvironment to mediate ocular fibrosis.
Ultimately, further understanding of the molecular patterns and biomechanics involved in conjunctival wound healing will allow for more targeted therapies to reduce IOP and prevent glaucoma-related complications.

3. Strategies to Prevent Ocular Fibrosis following Glaucoma Filtration Surgery

Although limiting intraoperative bleeding may attenuate fibroblast activity, this is insufficient to reduce long-term fibrosis. The following section discusses current and promising new therapies in development, which are summarized in Table 1.

3.1. Modulating Inflammation

Numerous anti-inflammatory agents including steroids and NSAIDs are used clinically to increase surgical success and modulate wound healing. Intraoperative and postoperative steroids can increase the success of glaucoma surgery [10,13]. Steroid use is mediated by the suppression of leukocyte concentration, distribution, and function. It ultimately diminishes the fibroblast activity in addition to the vascular effects. The reduction of neutrophils at the inflammatory site shifts lymphocytes, monocytes, and basophils out of circulation and into lymphoid tissue. Consequently, steroid inhibition of leukocyte concentration and function as well as vascular permeability led to less local tissue disruption, reduction in the amounts and activities of mitogens and growth factors, and decreased fibrin production, all of which result in attenuation of wound healing [13]. Topical steroids are often administered, but different dosages, routes, and durations of therapy have resulted in variable outcomes [104]. Topical corticosteroids use following trabeculectomy appears effective, but there has been no definitive consensus on dosing and duration of treatment [104]. The main challenges with steroid use are the systemic side effects, short aqueous half-life, prolonging its release, and the possibility of cataractogenesis with prolonged usage [105,106].
Although the usage of topical NSAIDs may enhance the lowering of IOP from topical prostaglandin analog treatment for glaucoma patients, the usage of NSAIDs with glaucoma filtration surgery remains controversial [107]. Clinically, NSAIDs are not routinely used after glaucoma filtration surgery. NSAIDs exhibit non-inferiority to steroids in terms of postoperative IOP control. A 2019 meta-analysis showed insufficient evidence to recommend one over the other [108]. Although most studies in the meta-analysis suggest no difference in the IOP-lowering ability of NSAIDs and steroids, other studies show conflicting evidence. A study by Yuen et al. compared topical dexamethasone to ketorolac after filtering glaucoma surgery with Ahmed glaucoma valve implantation. They found that mean IOP was significantly greater in the steroid group only at week 4 postoperatively, while the difference was not statistically significant at all other time points. In addition, NSAIDs showed a greater incidence of conjunctival retraction compared to dexamethasone [109]. However, a randomized controlled trial (RCT) by Breusegem et al. showed that topical ketorolac for 1 month preoperatively improved trabeculectomy outcomes when measured by the likelihood of postoperative needling. However, when ketorolac was compared to the steroid fluorometholone administered preoperatively, ketorolac was not as effective in reducing the need for additional postoperative IOP-lowering medications to reach the target IOP [57]. A retrospective cohort study showed that patients with filled NSAID prescriptions in the perioperative period had fewer interventions for bleb failure compared to patients without filled NSAID prescriptions [58]. These data suggest that steroids and NSAIDs may have differing effects on wound modulation following surgery, but our current understanding is that one is not necessarily superior to the other. Due to the limited and contradicting evidence, more investigation on this topic is necessary.

3.2. Modulating Would Healing

3.2.1. 5-FU

5-FU is a chemotherapeutic agent, and its antiproliferative effect stems from its ability to antagonize pyrimidine metabolism. Hence, DNA synthesis is inhibited, and cells die in the S-phase. 5-FU is effective at inhibiting human and animal fibroblasts in addition to inhibiting fibroblast-mediated collagen contraction. However, 5-FU is toxic to actively replicating tissues [39,79,110]. There is potential for the reduction of filtering surgical failure when injected through the conjunctiva postoperatively [40]; however, the increased number of visits, the discomfort of repeated injections, and epithelial toxicity are often cited as reasons why 5-FU is most commonly used on an ad hoc basis or in cases of imminent bleb failure [104]. In a study by Wormald et al., no evidence was found of an increased risk of serious sight-threatening complications, but common complications included hypotony maculopathy and wound leak [41].

3.2.2. MMC

MMC is a DNA-crosslinking-alkylating agent that inhibits the cell cycle, DNA replication, cell mitosis, and protein synthesis. MMC intercalates DNA and targets cells throughout the cell cycle to block cell proliferation. Many clinical studies have shown that MMC reduces scar formation, increases the rate of bleb survival, and maintains IOP reduction [42].
MMC is routinely used with subconjunctival/sub-Tenon’s glaucoma filtration surgery, such as trabeculectomy and deep sclerectomy. It is also used with newer subconjunctival MIGS procedures, including the XEN gel stent or Preserflo microshunt surgery, to prevent fibrosis and increase surgical success rates. Complications are often associated with antimetabolite treatment, including hypotony, blebitis, endophthalmitis, bleb leakage, and vision loss [111,112,113,114,115]. More detail on the innovations and history of MMC use has been provided previously [116].
Although the method of application may affect the rate of surgical failure and complications, multiple studies have found that it does not result in significant differences. In a study looking at Asian eyes, phacoemulsification-trabeculectomy combined with subconjunctival MMC injection had comparable outcomes to that of sponge-applied MMC, with a similar reduction in IOP at 1, 6, and 12 months postoperatively and a lower postoperative complication rate [43]. However, MMC injection may lower the need for clinic visits within the first 3 postoperative months and reduce the need for 5-FU intervention when compared to MMC sponge application [117]. In an RCT, Do et al. demonstrated that preoperative subconjunctival injection of MMC and intraoperative direct scleral application of MMC with surgical sponges resulted in comparable surgical outcomes [118]. However, some studies suggest that sub-Tenon injection of MMC produces more favorable bleb morphology after trabeculectomy [44,45,46]. Methods and dosage articles have been recently reviewed elsewhere by Bell et al. Due to the greater potency of MMC, intraoperative application of MMC has become popular with adjunctive postoperative 5-FU [42].
In an RCT, there were no differences in the types of complications from 5-FU and MMC, as both can contribute to bleb leakage, hypotony, choroidal effusion, and endophthalmitis in the short to medium term [119]. Though MMC inhibits fibroblast proliferation more effectively and permanently than 5-FU, it may be associated with higher rates of complications [80,81,82]. Experimental evidence and clinical use indicate that intraoperative MMC can cause pale, avascular blebs that can become thin and lead to epithelial surface breakdown. This progression is due to the MMC-induced apoptosis of conjunctival connective fibroblasts [120]. The toxic effects of MMC may be mitigated by combining a lower dosage of MMC with valproic acid, which has been demonstrated in a rabbit model [121,122]. There is no definite clinical recommendation for post-operative 5-FU as opposed to MMC.

3.2.3. Beta Radiation

In vitro studies have shown that beta radiation diminishes fibroblast proliferation, likely through a mechanism involving increased p53, a key enzyme in the cell cycle [83,84]. Kirwan et al. compared trabeculectomy with beta radiation to trabeculectomy without beta radiation across studies that included black African, Caucasian, and Chinese subjects and concluded the same findings in RCTs [47]. At the time of trabeculectomy, beta radiation can minimize scar tissue formation and increase the likelihood that surgery will effectively lower the IOP, but it has been shown to increase the risk of cataract formation [47]. In vitro studies have shown that beta radiation targets ECM production as opposed to fibroblast migration or contraction [84]. The advantages of beta radiation include ease of application, cost, and accessibility. Disadvantages of beta radiation include the possibility of cataract formation and keratopathy. In an RCT looking at trabeculectomies, beta radiation with trabeculectomy led to less bleb failure and reduced IOP compared with MMC with trabeculectomy in the South African population [123]. Another study in Egyptian eyes showed that preoperative beta radiation in addition to intraoperative MMC patients during trabeculectomy had greater IOP control. However, beta radiation may increase the risk of a cystic bleb. Beta radiation shows promise in increasing glaucoma filtration surgery success; however, there is not enough evidence to recommend beta radiation over current clinical standards of practice. Large, multicenter RCTs comparing beta radiation and MMC in glaucoma filtration surgery are warranted [48].
Overall, there is great variation in the usage of anti-scarring adjuncts before, during, and/or after surgery, and many adjuncts can be associated with a higher complication rate. A greater understanding of the cellular mechanisms of the wound healing response has led to the identification and modulation of potential new therapeutic targets.

3.2.4. Anti-TGFβ Agents

Monoclonal antibodies directed at TGFβ are advantageous due to their specificity. One of the key advantages of an antibody is its target specificity as opposed to a nonspecific mechanism of action, as in the case of 5-FU and MMC. Lerdelimumab is a monoclonal antibody to TGFβ2. Even though it was promising following laboratory studies, a human RCT failed to show the efficacy of Lerdelimumab in preventing the failure of trabeculectomy [124]. Fresolimnumab is a monoclonal antibody that neutralizes all forms of TGFβ; however, to our knowledge, it has not been tested in the setting of ocular fibrosis [85,86]. Further investigation into the use of monoclonal antibodies to TGFβ in glaucoma filtering surgery is warranted.
Targeting SMAD proteins by overexpressing inhibitory SMAD7 or reducing SMAD2, 3, or 4 can reduce stromal fibrosis [26]. In vivo studies have shown that expression of exogenous SMAD7 in burned corneal tissue results in reduced activation of SMAD signaling [125]. In work by Yamanaka et al., gene transfer of SMAD7 suppressed fibrogenic and inflammatory signals, including α-SMA, VEGF, and CTGF [59]. In work by Gupta et al., topical application of SMAD7 through recombinant adeno-associated virus serotype 5 to rabbit cornea post-photorefractive keratectomy resulted in a decrease in corneal fibrosis [126]. Topographical cues in the biophysical environment have also been shown to increase the expression of SMAD7 to prevent fibrosis by modulating TGFβ-induced myofibroblast differentiation and α-SMA expression [126]. To our knowledge, directly targeting SMAD proteins has not been studied in glaucoma filtration surgery, but this strategy may have therapeutic potential in the prevention of excess scarring.
Fibroblasts in the cornea express TGFβ induced factors (TGIF) 1 and 2. TGIFs repress SMAD-dependent signaling at the level of transcription. In vitro studies have demonstrated that TGIF1 gene editing via CRISPR/Cas9 can attenuate myofibroblast formation and protein levels of profibrotic genes [127]. Vorinostat is a histone deacetylase inhibitor that increases TGIF levels to inhibit TGFβ profibrotic expression of genes [91,92]. Vorinostat has been studied in animal models of trabeculectomy and is discussed in Section 3.2.12. Trichostatin and ITF2357 are other histone deacetylase inhibitors that can also affect SMAD signaling [93,94]. Trichostatin A has been shown to decrease bleb vascularity, leukocyte infiltration, and expression of α-SMA and TGF-β1 in the conjunctiva in rat models of trabeculectomy [128]. ITF2357 has been shown to attenuate corneal fibrosis in vivo but has not been studied in models of glaucoma filtration surgery.
SMAD-independent pathways may also be targeted through PPARγ ligands, p38 MAPK inhibitors, JNK inhibitors, mTOR (PI3K) inhibitors, and Rho signaling inhibitors. The PPARγ ligand Rosiglitazone effectively controls corneal fibrosis in vivo and in vitro, while pioglitazone has been shown to modulate fibrosis in vitro [30,129,130,131]. Studies have shown promise for p38 MAPK inhibitors in vitro and JNK inhibitors in a rat model [30,124,132]. The mTOR inhibitor Rapamycin has been shown to reduce corneal scarring in a rabbit model [133,134]. The Rho family of small GTPases orchestrate fundamental biological processes, including cell migration and actin cytoskeleton dynamics [135,136]. ROCKs are Rho effectors that play an important role in regulating stress fiber formation in fibroblasts and epithelial cells in vitro [137]. ROCK inhibitors have been found to attenuate fibroproliferation and scar formation in a rabbit model of glaucoma surgery, which may be due to decreased matrix contraction, thereby reducing scarring [136].
While TGFβ2 is the predominant isoform of TGFβ in the eye and the dominant isoform in conjunctival fibrosis, TGFβ1 is the dominant mediator of the corneal fibrotic response [138]. TGFβ1 is involved in ECM protein expression and pathological tissue fibrosis [139]. In vitro studies have shown that KCa3.1 (calcium-activated potassium channel) inhibitor TRAM34 reduced TGFβ-mediated cell migration and myofibroblast formation in human corneal fibroblasts [140]. CTGF, a regulatory molecule in the TGFβ pathway, is another target for mediating scarring, as it is involved with ECM production, actin cytoskeletal dynamics, and contractile force in human subconjunctival fibroblasts. A study by Lee et al. showed that CTGF suppression by CRISPR in a rabbit glaucoma filtration surgery model showed significantly better survival at the surgery site, less subconjunctival fibrosis, limited collagen deposition, and reduced cellularity when compared with untreated eyes [141].
Pirfenidone is another novel agent that has shown antifibrotic and antiangiogenic potential, which has been established by many in vitro and in vivo studies [63,95,96]. It is currently implicated in treating idiopathic pulmonary fibrosis [97]. It is possible that pirfenidone inhibits TGFβ and reduces TNFα, but the mechanism of action is unknown [142]. In vitro, pirfenidone inhibits extracellular matrix deposition in ocular fibroblasts and decreases fibroblast proliferation, migration, and contraction [143,144]. It has been shown that 0.5% pirfenidone eye drops improve trabeculectomy bleb success in a rabbit model [64].

3.2.5. Anti-YAP/TAZ

In vitro studies have found the Yes-associated protein (YAP)/transcriptional co-activator PDZ-binding motif (TAZ) to be involved in the activation of SMAD2/3. YAP/TAZ is a potential target in TGFβ2–mediated conjunctival fibrosis and TGFβ1-induced myofibroblast transformation [87,88]. YAP/TAZ interacts with SMAD proteins and likely transduces stiffness to the myofibroblast phenotype in the cornea [138]. Verteporfin is a YAP/TAZ inhibitor, and in vitro studies have found that it may improve outcomes in glaucoma surgery by suppressing TGFβ2-YAP/TAZ-SMAD signaling [87]. Therefore, YAP/TAZ may act as orchestrating molecules that integrate the biophysical and biochemical responses.
Other evolving approaches to targeting the TGFβ pathway include long non-coding RNAs, which is a strategy that silences gene expression. MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression at the post-transcriptional level by base-pairing with complementary sequences of the mRNA, which cleaves or destabilizes the mRNA. A study by Hwang et al. found that miRNA 143/1145 inhibition in human Tenon’s fibroblasts may be a promising target for subconjunctival fibrosis [60]. Overexpression of miRNA 26a and 26b may also be targeted, as they significantly inhibit lens fibrosis in vitro and in vivo [145].

3.2.6. Anti-SPARC

Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein involved in ECM production and organization that has been shown to be upregulated by TGFβ2 through a post-transcriptional mechanism [89,90]. Seet et al. showed that silencing SPARC in Tenon’s fibroblasts reduces profibrotic gene expression without stimulating apoptosis, as with MMC [146].
SPARC knock-out mice have shown improved surgical survival due to less collagenous ECM and smaller collagen fibril diameter [61]. To target this gene in humans, gene therapies involving siRNAs are in development. Nanoparticles serve as a vehicle to transport ligands such as antibodies and peptides into the cell for gene therapy. Nanoparticle-based siRNA delivery has been tested to target the SPARC gene and is still being tested in animal studies; pilot studies have shown that this method is safe and feasible [62]. Another delivery system of SPARC siRNA utilizing a hydrogel is currently in animal studies. Its safety profile is promising and shows effective inhibition of subconjunctival scarring in a rabbit model [147].

3.2.7. Anti-PDGF Subunit B Agents

Studies suggest that E10030, an anti-PDGF aptamer, inhibits angiogenesis and tumor growth, but its effect in the context of ocular fibrosis has not been well-studied [65,66]. A study on aptamers ARC126 and ARC127, aptamers that bind PDGF subunit B, showed that there is potential for aptamers in reducing scarring and fibroblast proliferation in mouse models with proliferative retinopathies [148].
Kinase inhibitors such as Nintedanib inhibit the PDGF receptor, VEGF receptor, and FGF receptor tyrosine kinases. They reduce the TGFβ1-induced phosphorylation of SMAD2/3, p38 MAPK, and ERK1/2 to prevent Tenon’s fibroblast activation in vitro [97,149]. Thus, there are potent antifibrotic effects on Tenon’s fibroblasts, and these are potential therapeutic targets.

3.2.8. Anti-VEGF Agents

Angiogenesis plays an important role in wound healing, and VEGF plays a key role in the formation of pathological blood vessels. One study showed that VEGF is expressed in aqueous humor from glaucoma patients and rabbits that have undergone surgery [67]. VEGF was also found to stimulate the proliferation of Tenon’s fibroblasts in cell cultures. In vitro, bevacizumab has been shown to inhibit fibroblast proliferation and improve scarring in an animal model by reducing angiogenesis and collagen deposition [67]. Clinical studies on IOP after using subconjunctival bevacizumab injection in glaucoma patients show equal effectiveness in IOP reduction compared with MMC and potentially a better safety profile, but bevacizumab delivered by sponge had no advantage over MMC [150,151]. Intracameral bevacizumab and MMC in trabeculectomy were shown by another group to have differences in surgical success [152]. Additionally, the anti-scarring effect of combined bevacizumab and MMC was not significantly different from MMC alone, but bevacizumab and 5-FU led to less scarring compared to either agent alone both in vitro and in vivo [153,154]. Intravitreal ranibizumab was also shown to lead to less vascularity with more diffuse blebs when compared to MMC [155]. However, there is still controversy regarding the use of anti-VEGF agents, as some studies show that there is insufficient evidence for ranibizumab and bevacizumab injection use in glaucoma surgery [145,156].

3.2.9. Fibrinolytic Therapy

Tissue plasminogen activator (tPA) has been deemed useful in dissolving fibrin in blood clots after glaucoma surgery. However, hyphema, profound hypotony, and anterior chamber flattening were found in 11% of patients, though no long-term complications were associated with intracameral tPA use after a mean follow-up of 2.5 years [49]. Moreover, an RCT by Barequet et al. showed that sponge application of MMC during trabeculectomy with intracameral tPA may increase trabeculectomy success rates without a difference in safety compared to sponge application of MMC alone [157]. Urokinase is a thrombolytic agent that has been shown by one study to reduce IOP after glaucoma surgery without adverse effects when injected into the anterior chamber. However, the long-term safety of intracameral injection is uncertain [158].

3.2.10. Collagen-Platelet Interaction Inhibitors

Saratin is a 12kD protein that was isolated from the saliva of the leech Hirudo medicinalis. It interferes with platelet integrin α2β1-collagen and von Willebrand factor-collagen binding, preventing platelet aggregation in response to injury [159]. Because of the interference of platelet aggregation, saratin limits PDGF, TGFβ, IGF, and EGF [99]. A single intraoperative topical application of saratin did not outperform MMC but a combination of intraoperative topical application with two additional postoperative injections did not cause bleb avascularity and tissue thinning, which is often associated with MMC treatment [68]. Although Saratin is not used clinically, further exploration into its dosing is warranted.

3.2.11. LOX and LOXL Antibodies

The lysyl oxidases (LOX) are a family of enzymes that plays a role in the crosslinking of collagen and elastin [160]. Lysyl oxidase-like 2 (LOXL2) is reported to be involved in many fibrotic diseases, including lung and myocardial fibrosis [161,162]. There is also a correlation between increased LOXL2 and TGFβ levels [163]. A murine model by Adachi et al. found that LOX genes are overexpressed in the bleb region in glaucoma filtration surgery [164]. In a rabbit model of trabeculectomy, targeting LOXL2 with an inhibitory monoclonal antibody reduced pathologic angiogenesis, inflammation, and fibrosis [69].

3.2.12. MMP Inhibitors

MMPs are proteolytic enzymes that are essential in all phases of wound healing [70]. They have an important role in wound remodeling–degrading extracellular matrix components while also being able to synthesize collagen and the extracellular matrix. MMPs are present in the bleb walls of Molteno implants of human eyes [165]. In vitro, MMP inhibition has been shown to reduce collagen contraction, cell migration, and collagen production without cellular toxicity [100]. Rabbits undergoing glaucoma filtration surgery treated with ilomastat, an MMP inhibitor, resulted in increased bleb survival, significantly lowered IOP over 30 days, and less scar tissue histologically compared to rabbits treated with the vehicle phosphate-buffered saline [70]. Topical doxycycline, another MMP inhibitor, was found to be similarly effective compared to intraoperative MMC in an in vivo rabbit glaucoma filtering surgery bleb survival study [166].
More recent preclinical tests in an animal model showed reduced inflammation and extracellular matrix remodeling with subconjunctival iIlomastat injection [71]. Preliminary results from a rabbit model show that adequate conjunctival tissue penetration and therapeutic concentrations within the sclera and conjunctiva and aqueous humor can be achieved [167].

3.2.13. MRTF/SRF Inhibitors

There is increasing evidence that the serum response factor (SRF) and myocardin-related transcription factor (MRTF) play an essential role in the activation of fibroblasts [168]. The MRTF-A/SRF transcription pathway is an important upstream regulator of MMP expression in ocular fibrosis [72,169]. Because of this, the MRTF/SRF pathway represents a potential novel target to inhibit MMP expression [72,168,169].
Nanoparticle-based siRNAs have also been in development as a method to induce MRTF silencing. In vitro studies of nanoparticle-based siRNAs showed silenced expression of the MRTF-B gene in human Tenon’s fibroblasts and blocked collagen matrix contraction while not being cytotoxic [101]. In vivo studies of nanoparticle-based siRNAs doubled bleb survival and decreased conjunctival scarring, with no adverse side effects, in a rabbit model of glaucoma filtration surgery [72].

3.2.14. Epigenetic Modifiers

Epigenetic modifiers regulate gene expression through methylation and acetylation of DNA and histone proteins. By altering DNA structure, histone modifiers regulate how DNA binds to its transcription factors [170]. Suberoylanilide hydroxamic acid (SAHA/vorinostat) is an HDAC inhibitor currently approved by the FDA for the treatment of cutaneous T-cell lymphoma. SAHA has been shown to improve outcomes with a pre-op subconjunctival application in a rabbit model [73]. On the other hand, a single intraoperative injection of SAHA in a rabbit model did not prolong bleb survival [171]. HDAC inhibition is another potential novel agent that can modulate glaucoma filtration surgery wound healing without the adverse effects associated with 5-FU and MMC; more investigation into its mechanism and clinical studies are necessary.

3.2.15. Cell Cycle Targets

Methods of gene therapy involve increasing expressions of inhibitory genes and proteins in the cell cycle. Human p53 is a tumor suppressor gene that plays a key role in arresting cell cycle progression to allow DNA repair or inducing apoptosis if the damage is too extensive [172]. Many studies have demonstrated p53 as an active player in human Tenon’s fibroblast migration and growth [102,103]. Using a recombinant adenovirus for p53, Johnson et al. induced overexpression of p53 in human Tenon’s fibroblasts and significantly inhibited fibroblast proliferation and DNA synthesis [173].
Perkins et al. placed one of p53′s downstream effectors, p21, in a similar recombinant adenovirus vector for use in rabbit trabeculectomy. They found that eyes undergoing p21 gene therapy performed similarly to MMC in terms of decreasing IOP and preventing the proliferation of fibrosis [74]. One consideration with the use of viral vectors is the rare but inherent risk of mutagenesis. At least two cases of lymphoproliferative disorders related to gene therapy have been described in the literature.
There is also potential for therapies that target other components of the cellular division pathway, including CDKN1B. In a rabbit model of glaucoma filtration surgery, Yang et al. found that overexpression of CDKN1B, a CDK inhibitor, may reduce the severity of scar formation and improve surgical outcomes by reducing fibroblast proliferation [75].

3.2.16. Broad-Spectrum Immunosuppressives

Cyclosporine is an immunosuppressive agent that prevents T-cell activation and subsequent T-cell mediated inflammation by reducing IL-2 and IL-2 receptor expression, though its benefit to glaucoma surgery outcomes is still uncertain as there is conflicting evidence. An in vivo study by Park et al. showed that postoperative topical 2% cyclosporine can enhance the effectiveness of glaucoma drainage implant surgery measured by IOP and improve the flow resistance through the implant capsule, although there was no difference in fibroblast density [76]. Recent in vivo studies with different delivery methods and concentrations have supported this data [174,175]. However, 2% cyclosporine intraoperatively and postoperatively was not associated with IOP or prolonged bleb survival in rabbits when compared with MMC [176]. In human clinical trials, topical 0.05% cyclosporine was shown to have no effect on postoperative bleb function or IOP following trabeculectomy, although there was a decrease in ocular surface disease [177]. Sirolimus is a macrolide that is used as an immunomodulatory medication that decreases the response of B and T cells and reduces the response of cytokines such as IL-2. Sirolimus applied via a poly(lactic-co-glycolic acid) sustained delivery film in a rabbit model was efficacious at preventing filtration bleb scarring and increasing the success rate of filtration surgery [178].

3.3. Modulating Mechanotransduction

Over the last two decades, adjuncts to conventional glaucoma surgeries have been studied to improve success rates. Because mechanotransduction initiates biochemical responses, modulating mechanical stress is imperative to maintaining the subconjunctival/sub-Tenon’s space after surgery. Physical spacers separating the subconjunctival and episcleral tissues may be used to prevent fibrosis and are still under study, as early contact between the two surfaces may lead to fibrosis and result in flat and nonfunctional blebs.

3.3.1. Human Amniotic Membrane

The human amniotic membrane may be beneficial due to its anti-inflammatory, antifibrotic, and antiangiogenic properties [179]. It is integrated into the host as a surgical graft or temporarily used as a biological bandage. Two groups have shown that in vivo amniotic membrane transplantation (AMT) in the construction of filtering blebs attenuates the healing response. However, the improvement in bleb survival must be weighed against the association with delayed healing [77]. Sheha et al. compared trabeculectomy with MMC to trabeculectomy with MMC and AMT. They found that the latter group had higher success rates, lower postoperative IOPs, and fewer complications. However, a 2021 study by Roque et al. found that trabeculectomy combined with MMC and AMT did not show better results than trabeculectomy with MMC alone [180]. A 2015 Cochrane Review found 18 studies investigating the use of AMT with trabeculectomy. The review noted the data as low-quality but concluded that AMT can improve IOP slightly and may lead to fewer complications [181]. A systematic review of five RCTs showed that mean IOP was lower and that complications, including a flat anterior chamber and hyphema, were decreased in trabeculectomy with AMT compared to trabeculectomy without AMT [182].

3.3.2. Perfluoropropane Gas

Perfluoropropane (C3F8) expansile gas has promise in acting as a spacer in the augmentation of glaucoma-filtering blebs. It can be injected into the subconjunctival space at the end of surgery and lasts for 2–3 weeks. Animal studies and pilot human studies have shown safe and promising results in the early post-op period as well as long-term IOP control [50]. Thus, injection of C3F8 gas at the time of glaucoma filtering surgery may be a promising technique for increasing bleb survival and reducing fibrosis [183].

3.3.3. Sodium Hyaluronate

Healaflow is a cross-linked sodium hyaluronate that is absorbed slowly to prevent scarring, though it is not currently available in the US. Multiple human clinical studies have shown that Healaflow is a safe adjunct in both trabeculectomy and deep sclerectomy, and some studies have found that Healaflow enhances efficacy compared to these procedures without Healaflow [48,51,52,53,54,55]. However, in an RCT, Mudhol et al. showed that Healaflow and low-dose MMC are equally efficacious [52].

3.3.4. Collagen Matrix

Ologen is a biodegradable three-dimensional collagen-glycosaminoglycan copolymer matrix implant that may also act as a physical spacer [56]. In an RCT, Ologen was shown to improve IOP control and blunt the hypertensive phase in Ahmed glaucoma valve surgery immediately after surgery but not at 6 or 12 months post-surgery [184]. Although early clinical studies comparing the rates of complications and efficacy of Ologen to MMC in trabeculectomy showed mixed results, recent trials have shown the comparable or superior efficacy of Ologen [56]. When comparing Ologen and perfluoropropane in trabeculectomy, the IOP values were similar in the first 12 months of follow-up, but the Ologen group showed a more significant reduction in IOP during the last 24 months of follow-up [50].

3.4. Modulating the Hypertensive Phase

The hypertensive phase, an increase in IOP that occurs usually in the first month postoperatively, is common following glaucoma filtration surgery. A higher preoperative IOP and younger age are risk factors for the hypertensive phase [185,186]. Pharmacological agents aimed to decrease the occurrence of the hypertensive phase include prostaglandin analogs and aqueous suppressant treatment [187]. However, recent findings suggest that the use of aqueous suppressants and prostaglandin analogs may affect wound healing. Aqueous suppressants have been shown to decrease the expression of α-SMA, reduce the transformation of fibroblasts to myofibroblasts, attenuate growth factors, and reduce mechanical stress from aqueous pressure on the bleb [187]. Prostaglandin analogs have been shown to induce collagen contraction, upregulate IL-2, and upregulate MMP-9. Jung et al. also showed that early treatment with aqueous suppressants decreased fibrosis in the bleb but early treatment with prostaglandin analogs did not decrease fibrosis. IOP at 4 weeks post-op was also lower in the aqueous suppressant group.
In current clinical practice, the most commonly used agents to prevent fibrosis and improve surgical outcomes are corticosteroids and antimetabolites. Most glaucoma specialists in the United States utilize MMC at the time of glaucoma filtration surgery (e.g., trabeculectomy), either via subconjunctival injection or soaked pledgets. The concentration of MMC and length of application of MMC varies by surgeon and patient characteristics. 5-FU as an antifibrotic is more commonly used intraoperatively in the United Kingdom. Postoperatively, topical corticosteroids are the mainstay of treatment to suppress ocular fibrosis. Typically, surgeons will prescribe topical prednisolone acetate 1% every 2 h while awake (8 times per day) or difluprednate 0.05% every 4 h while awake (4 times per day) for the first week and sometimes the first month after surgery before beginning to taper. Most surgeons do not utilize NSAIDs to mitigate fibrosis after glaucoma filtration surgery.
Overall, there is a great need for more advancements in our understanding of emerging and repurposed treatments that target fibrosis. Further investigation into solutions that will carry these molecular techniques from bench to bedside will allow for the ultimate goal of modulating fibrosis in glaucoma surgery, which will prevent vision loss among millions of people worldwide.

4. Novel Delivery Techniques and Biomaterials

Delivery methods of antifibrotic agents are important due to their rapid clearance from the subconjunctival space. It is important to prevent clearance while also ensuring that additional material–tissue interactions and localized foreign body responses are minimized. To prevent rapid clearance of antifibrotics, sustained-release implants, sponges, and injections have been used in certain therapies.
Hydrogels are a network of cross-linked polymer chains that are highly absorbent [188]. They are capable of increasing drug residence time and sustaining drug delivery to biomechanically modulate ocular wound healing. Many drugs with the potential for preventing ocular fibrosis have been tested in hydrogels for sustained release, including 5-FU, MMC, ranibizumab, and dexamethasone [115,188,189,190,191]. Hydrogels can provide specific mechanical signaling cues by acting as scaffolds for the infiltrating restorative cells, which makes them a promising approach for spatiotemporal control of ocular fibrosis.
Liposomal delivery systems have also been developed as a method of prolonging drug levels in the eye. Drug levels of 5-FU and mitoxantrone have been studied in rabbit models of glaucoma surgery, which have shown increased conjunctival drug concentrations and reduced ocular side effects [192,193].
Encapsulation of therapies in nanoparticles can enhance permeation across cell membranes and prevent enzymatic degradation [193]. Recent studies have shown that continuous nanoparticle drug delivery to targeted tissues and cells can be more effective than traditional drug delivery methods [194,195]. LDL receptors are overexpressed in activated Tenon’s fibroblasts. LDL-MMC nanoparticles specifically bind to upregulate LDL receptors on activated fibroblasts and are a novel therapy due to their selective targeting, small drug dose requirement, better bioavailability, and reduced cellular toxicity [196].
Alternative modes of drug delivery have been studied to increase the specificity of drug targets. Implanted biomaterials initiate nonspecific protein adsorption to the material surface, forming a matrix that is conducive to cellular adhesion. Biomaterial physical properties such as size, surface topography, and porosity influence fibroblast behavior and are critical to maintaining longevity and surgical efficacy [197].
There are many novel implants with biomaterials that are being studied in animal models that are promising for increasing the efficacy of glaucoma surgery. Electrospinning allows for the incorporation of polymers into nanofibers that can be manipulated into various combinations. Nano-structured glaucoma shunts are promising and have been tested in a rabbit model; they showed immediate IOP reduction and prevented post-op hypotony [198].
Dendrimers, hyperbranched macromolecules, can be engineered to a precise structure. They can be synthesized with anti-inflammatory properties. In vitro experiments with dendrimers developed by Shaunak et al. were found to reduce synthesis of pro-inflammatory chemokines and inhibit angiogenesis, in addition to increasing glaucoma filtration surgery success, in a rabbit model filtration surgery [199].
A custom-tailored expanded polytetrafluoroethylene (ePTFE) glaucoma drainage implant is another novel approach. The ePTFE glaucoma drainage implant is promising due to its high biocompatibility and inertness. It was found to reduce capsule thickness in blebs and increase their permeability compared with silicone implants in rabbits [199,200]. Collagen deposition is substantially lower with the ePTFE device than with the silicon implant control after one to three months. This device is currently being studied in human clinical trials, and the data are planned for release in 2023 [98].
Poly(styrene-block-isobutylene-block-styrene) (SIBS) is a stable, conforming, and biologically inert biomaterial that has been combined with the glaucoma drainage microtube as the PreserFlo® MicroShunt [201]. SIBS results in virtually no foreign body reaction, which translates to insignificant inflammation and capsule formation. SIBS is still in clinical studies but shows promising efficacy and safety outcomes, although there is not yet enough evidence to recommend it over the current guidelines.
The Beacon Aqueous Microshunt shunts aqueous humor directly to the tear film [202]. Its channel is composed of polymerized, superhydrophilic polyethylene glycol, which prevents protein adsorption and bacterial attachments. This device is still in clinical trials.
In summary, various delivery techniques may significantly improve the glaucoma filtration surgery success rate. Many of these avenues of possibility in new drug delivery systems have not been fully characterized but have the potential for fine-tuning optimal drug delivery times, reducing cellular toxicity, and increasing the precision of drug delivery. Prolonging the presence of antifibrotic agents and utilizing novel biomaterials that attenuate the inflammatory response have significant implications in the treatment of glaucoma, allowing for greater IOP control and reducing post-op complications.

5. Conclusions

Several agents, devices, and biomaterials are potential alternatives to the current anti-fibrotic therapies, but they remain unavailable for clinical use. Many of the methods for the prevention of subconjunctival and sub-Tenon’s fibrosis have not been tested in large preclinical in vivo animal models and human RCTs. Retrospective case series and nonrandomized prospective studies provide insight into the potential for new glaucoma procedures and glaucoma filtration surgery adjuncts, but these early results should be interpreted with caution. Relevant evidence for the potential of new biomaterials and gene therapies is limited but is accumulating, as many are still in the early stages of testing. Likewise, several novel drug delivery approaches for antifibrotics are promising, but translation of these technologies to the bedside can be challenging. This review has described potential therapeutic targets for the prevention of ocular fibrosis after traditional incisional glaucoma filtering surgery, but RCTs are needed to improve our understanding of the long-term efficacy and safety of these novel therapies and to further define their role in clinical practice.

Author Contributions

Conceptualization, R.R.M. and A.D.W.; writing—original draft preparation, C.G.S.; writing—review and editing, C.G.S., N.R.S., R.R.M. and A.D.W.; figures, N.R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funding in part by the United States Department of Veterans Health Affairs, grant numbers 1I01BX00357 and IK6BX005646, and by NIH/NEI, grant number R01EY0343319 (RRM).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-FUFluorouracil
AMTAmniotic Membrane Transplantation
AAVAdeno-Associated Virus
α-SMAalpha-Smooth Muscle Actin
C3F8Perfluoropropane
COL-GAGCollagen Glycosaminoglycan
CTGFConnective Tissue Growth Factor
ECMExtracellular Matrix
ePTFEExpanded Polytetrafluoroethylene
IOPIntraocular Pressure
LOXL2Lysyl Oxidase-Like 2
MMCMitomyosin C
MMPMatrix Metalloproteinase
MRTFMyocardin-related transcription factor
POPost-operative
PDGFPlatelet-Derived Growth Factor
PTKPhototherapeutic Keratectomy
RCTsRandomized Controlled Trials
SAHASuberoylanilide Hydroxamic Acid
SC Inj.Subconjunctival Injection
SIBSPoly(styrene-block-isobutylene-block-styrene)
SMADSmall Mothers Against Decapentaplegic
SPARCSecreted Protein Acidic and Rich in Cysteine
SRFSerum Response Factor
ST Inj.Subtenon Injection
TGFβTransforming Growth Factor beta
TGIFTGF-β induced factors
tPATissue plasminogen activator
VEGFVascular Endothelial Growth Factor
YAP/TAZYes-associated protein/ transcriptional co-activator PDZ-binding motif

References

  1. Yu-Wai-Man, C.; Treisman, R.; Bailly, M.; Khaw, P.T. The Role of the MRTF-A/SRF Pathway in Ocular Fibrosis. Investig. Ophthalmol. Vis. Sci. 2014, 55, 4560–4567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Yan, C.; Boyd, D.D. Regulation of matrix metalloproteinase gene expression. J. Cell. Physiol. 2007, 211, 19–26. [Google Scholar] [CrossRef] [PubMed]
  3. Jung, K.I.; Woo, J.E.; Park, C.K. Effects of aqueous suppressants and prostaglandin analogues on early wound healing after glaucoma implant surgery. Sci. Rep. 2019, 9, 5251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Parikh, K.S.; Josyula, A.; Omiadze, R.; Ahn, J.Y.; Ha, Y.; Ensign, L.M.; Hanes, J.; Pitha, I. Nano-structured glaucoma drainage implant safely and significantly reduces intraocular pressure in rabbits via post-operative outflow modulation. Sci. Rep. 2020, 10, 12911. [Google Scholar] [CrossRef]
  5. Flaxman, S.R.; Bourne, R.R.A.; Resnikoff, S.; Ackland, P.; Braithwaite, T.; Cicinelli, M.V.; Das, A.; Jonas, J.B.; Keeffe, J.; Kempen, J.H.; et al. Global causes of blindness and distance vision impairment 1990–2020: A systematic review and meta-analysis. Lancet Glob. Health 2017, 5, e1221–e1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Jonas, J.B.; Aung, T.; Bourne, R.R.; Bron, A.M.; Ritch, R.; Panda-Jonas, S. Glaucoma. Lancet 2017, 390, 2183–2193. [Google Scholar] [CrossRef]
  7. Zhang, N.; Wang, J.; Li, Y.; Jiang, B. Prevalence of primary open angle glaucoma in the last 20 years: A meta-analysis and systematic review. Sci. Rep. 2021, 11, 13762. [Google Scholar] [CrossRef]
  8. Conlon, R.; Saheb, H.; Ahmed, I.I.K. Glaucoma treatment trends: A review. Can. J. Ophthalmol. 2017, 52, 114–124. [Google Scholar] [CrossRef]
  9. Lusthaus, J.; Goldberg, I. Current management of glaucoma. Med. J. Aust. 2019, 210, 180–187. [Google Scholar] [CrossRef]
  10. Van Bergen, T.; Van de Velde, S.; Vandewalle, E.; Moons, L.; Stalmans, I. Improving patient outcomes following glaucoma surgery: State of the art and future perspectives. Clin. Ophthalmol. 2014, 8, 857–867. [Google Scholar] [CrossRef] [Green Version]
  11. Holló, G. Wound Healing and Glaucoma Surgery: Modulating the Scarring Process with Conventional Antimetabolites and New Molecules. Dev. Ophthalmol. 2017, 59, 80–89. [Google Scholar] [CrossRef]
  12. Skuta, G.L.; Parrish, R.K. Wound healing in glaucoma filtering surgery. Surv. Ophthalmol. 1987, 32, 149–170. [Google Scholar] [CrossRef] [Green Version]
  13. Lama, P.J.; Fechtner, R.D. Antifibrotics and Wound Healing in Glaucoma Surgery. Surv. Ophthalmol. 2003, 48, 314–346. [Google Scholar] [CrossRef]
  14. Shu, D.Y.; Lovicu, F.J. Myofibroblast transdifferentiation: The dark force in ocular wound healing and fibrosis. Prog. Retin. Eye Res. 2017, 60, 44–65. [Google Scholar] [CrossRef]
  15. Zada, M.; Pattamatta, U.; White, A. Modulation of Fibroblasts in Conjunctival Wound Healing. Ophthalmology 2018, 125, 179–192. [Google Scholar] [CrossRef]
  16. Schlunck, G.; Meyer-Ter-Vehn, T.; Klink, T.; Grehn, F. Conjunctival fibrosis following filtering glaucoma surgery. Exp. Eye Res. 2016, 142, 76–82. [Google Scholar] [CrossRef]
  17. Pakyari, M.; Farrokhi, A.; Maharlooei, M.K.; Ghahary, A. Critical Role of Transforming Growth Factor Beta in Different Phases of Wound Healing. Adv. Wound Care 2013, 2, 215–224. [Google Scholar] [CrossRef] [Green Version]
  18. Pierce, G.F.; A Mustoe, T.; Lingelbach, J.; Masakowski, V.R.; Griffin, G.L.; Senior, R.M.; Deuel, T.F. Platelet-derived growth factor and transforming growth factor-beta enhance tissue repair activities by unique mechanisms. J. Cell Biol. 1989, 109, 429–440. [Google Scholar] [CrossRef]
  19. Pasquale, L.R.; Dorman-Pease, M.E.; Lutty, G.A.; Quigley, H.A.; Jampel, H.D. Immunolocalization of TGF-beta 1, TGF-beta 2, and TGF-beta 3 in the anterior segment of the human eye. Investig. Ophthalmol. Vis. Sci. 1993, 34, 23–30. [Google Scholar]
  20. Granstein, R.D.; Staszewski, R.; Knisely, T.L.; Zeira, E.; Nazareno, R.; Latina, M.; Albert, D.M. Aqueous humor contains transforming growth factor-beta and a small (less than 3500 daltons) inhibitor of thymocyte proliferation. J. Immunol. 1990, 144, 3021–3027. [Google Scholar] [CrossRef]
  21. Tovar-Vidales, T.; Clark, A.F.; Wordinger, R.J. Transforming growth factor-beta2 utilizes the canonical Smad-signaling pathway to regulate tissue transglutaminase expression in human trabecular meshwork cells. Exp. Eye Res. 2011, 93, 442–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Khaw, P.T.; Occleston, N.L.; Schultz, G.; Grierson, I.; Sherwood, M.B.; Larkin, G. Activation and suppression of fibroblast function. Eye 1994, 8, 188–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Roberts, A.B.; Sporn, M.B.; Assoian, R.K.; Smith, J.M.; Roche, N.S.; Wakefield, L.M.; I Heine, U.; Liotta, L.A.; Falanga, V.; Kehrl, J.H. Transforming growth factor type beta: Rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc. Natl. Acad. Sci. USA 1986, 83, 4167–4171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Stahnke, T.; Gajda-Deryło, B.; Jünemann, A.G.; Stachs, O.; Sterenczak, K.A.; Rejdak, R.; Beck, J.; Schütz, E.; Möller, S.; Barrantes, I.; et al. Suppression of the TGF-β pathway by a macrolide antibiotic decreases fibrotic responses by ocular fibroblasts in vitro. R. Soc. Open Sci. 2020, 7, 200441. [Google Scholar] [CrossRef] [PubMed]
  25. Derynck, R.; Budi, E.H. Specificity, versatility, and control of TGF-β family signaling. Sci. Signal. 2019, 12, eaav5183. [Google Scholar] [CrossRef] [Green Version]
  26. Kamil, S.; Mohan, R.R. Corneal stromal wound healing: Major regulators and therapeutic targets. Ocul. Surf. 2021, 19, 290–306. [Google Scholar] [CrossRef]
  27. Massagué, J. NEW EMBO MEMBERS REVIEW: Transcriptional control by the TGF-beta/Smad signaling system. EMBO J. 2000, 19, 1745–1754. [Google Scholar] [CrossRef] [Green Version]
  28. Moustakas, A.; Pardali, K.; Gaal, A.; Heldin, C.-H. Mechanisms of TGF-β signaling in regulation of cell growth and differentiation. Immunol. Lett. 2002, 82, 85–91. [Google Scholar] [CrossRef]
  29. Hayashi, H.; Abdollah, S.; Qiu, Y.; Cai, J.; Xu, Y.-Y.; Grinnell, B.W.; Richardson, M.A., Jr.; Topper, J.N.; Gimbrone, M.A.; Wrana, J.L.; et al. The MAD-Related Protein Smad7 Associates with the TGFβ Receptor and Functions as an Antagonist of TGFβ Signaling. Cell 1997, 89, 1165–1173. [Google Scholar] [CrossRef] [Green Version]
  30. Shi, L.; Chang, Y.; Yang, Y.; Zhang, Y.; Yu, F.-S.X.; Wu, X. Activation of JNK Signaling Mediates Connective Tissue Growth Factor Expression and Scar Formation in Corneal Wound Healing. PLoS ONE 2012, 7, e32128. [Google Scholar] [CrossRef] [Green Version]
  31. Jester, J.V.; Ho-Chang, J. Modulation of cultured corneal keratocyte phenotype by growth factors/cytokines control in vitro contractility and extracellular matrix contraction. Exp. Eye Res. 2003, 77, 581–592. [Google Scholar] [CrossRef]
  32. Maltseva, O.; Folger, P.; Zekaria, D.; Petridou, S.; Masur, S.K. Fibroblast growth factor reversal of the corneal myofibroblast phenotype. Investig. Ophthalmol. Vis. Sci. 2001, 42, 2490–2495. [Google Scholar]
  33. Tomasek, J.J.; Gabbiani, G.; Hinz, B.; Chaponnier, C.; Brown, R.A. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat. Rev. Mol. Cell Biol. 2002, 3, 349–363. [Google Scholar] [CrossRef]
  34. Ingber, D.E. Cellular mechanotransduction: Putting all the pieces together again. FASEB J. 2006, 20, 811–827. [Google Scholar] [CrossRef]
  35. Orr, A.W.; Helmke, B.P.; Blackman, B.R.; Schwartz, M.A. Mechanisms of Mechanotransduction. Dev. Cell 2006, 10, 11–20. [Google Scholar] [CrossRef] [Green Version]
  36. A Kadri, O.; Wilcox, M.J. Surface tension controls capsule thickness and collagen orientation in glaucoma shunt devices. Biomed. Sci. Instrum. 2001, 37, 257–262. [Google Scholar]
  37. Raghunathan, V.K.; Thomasy, S.M.; Strøm, P.; Yañez-Soto, B.; Garland, S.P.; Sermeno, J.; Reilly, C.M.; Murphy, C.J. Tissue and cellular biomechanics during corneal wound injury and repair. Acta Biomater. 2017, 58, 291–301. [Google Scholar] [CrossRef]
  38. Pedersen, J.A.; Lichter, S.; Swartz, M.A. Cells in 3D matrices under interstitial flow: Effects of extracellular matrix alignment on cell shear stress and drag forces. J. Biomech. 2010, 43, 900–905. [Google Scholar] [CrossRef]
  39. Nakano, Y.; Araie, M.; Shirato, S. Effect of postoperative subconjunctival 5-fluorourcil injections on the surgical outcome of trabeculectomy in the Japanese. Graefe’s Arch. Clin. Exp. Ophthalmol. 1989, 227, 569–574. [Google Scholar] [CrossRef]
  40. Fluorouracil Filtering Surgery Study Group. The Five-year Follow-up of the Fluorouracil Filtering Surgery Study. Am. J. Ophthalmol. 1996, 121, 349–366. [Google Scholar] [CrossRef]
  41. Wilkins, M.R.; Bunce, C.; Wilkins, M. Post-operative 5-fluorouracil for glaucoma surgery. Cochrane Database Syst. Rev. 2001, 10, CD001132. [Google Scholar] [CrossRef]
  42. Bell, K.; Bezerra, B.D.P.S.; Mofokeng, M.; Montesano, G.; Nongpiur, M.E.; Marti, M.V.; Lawlor, M. Learning from the past: Mitomycin C use in trabeculectomy and its application in bleb-forming minimally invasive glaucoma surgery. Surv. Ophthalmol. 2021, 66, 109–123. [Google Scholar] [CrossRef] [PubMed]
  43. Ang, B.C.H.; Chiew, W.; Guo, X.; Lim, A.P.H.; Yip, L.W.L. Comparison of surgical outcomes of sponge application versus subconjunctival injection of Mitomycin-C during combined phacoemulsification and trabeculectomy surgery in Asian eyes. J. Curr. Ophthalmol. 2021, 33, 253. [Google Scholar] [CrossRef] [PubMed]
  44. Kandarakis, S.A.; Papakonstantinou, E.; Petrou, P.; Diagourtas, A.; Ifantides, C.; Georgalas, I.; Serle, J. One-Year Randomized Comparison of Safety and Efficacy of Trabeculectomy with Mitomycin C Sub-Tenon Injection versus Mitomycin C-Infused Sponges. Ophthalmol. Glaucoma 2022, 5, 77–84. [Google Scholar] [CrossRef] [PubMed]
  45. Esfandiari, H.; Pakravan, M.; Yazdani, S.; Doozandeh, A.; Yaseri, M.; Conner, I.P. Treatment Outcomes of Mitomycin C-Augmented Trabeculectomy, Sub-Tenon Injection versus Soaked Sponges, after 3 Years of Follow-up. Ophthalmol. Glaucoma 2018, 1, 66–74. [Google Scholar] [CrossRef]
  46. Pakravan, M.; Esfandiari, H.; Yazdani, S.; Douzandeh, A.; Amouhashemi, N.; Yaseri, M.; Pakravan, P. Mitomycin C-augmented trabeculectomy: Subtenon injection versus soaked sponges: A randomised clinical trial. Br. J. Ophthalmol. 2017, 101, 1275–1280. [Google Scholar] [CrossRef]
  47. Kirwan, J.F.; Rennie, C.; Evans, J.R. Beta radiation for glaucoma surgery. Cochrane Database Syst. Rev. 2012, 2012, CD003433. [Google Scholar] [CrossRef] [Green Version]
  48. El Mazar, H.M.; Mandour, S.S.; Mostafa, M.I.; Elmorsy, O.A. Augmented Subscleral Trabeculectomy with Beta Radiation and Mitomycin C in Egyptian Glaucoma Patients. Eur. J. Gastroenterol. Hepatol. 2019, 28, 637–642. [Google Scholar] [CrossRef]
  49. Slabaugh, M.; Salim, S. Use of Anti-VEGF Agents in Glaucoma Surgery. J. Ophthalmol. 2017, 2017, 1645269. [Google Scholar] [CrossRef]
  50. Shen, T.-Y.; Hu, W.-N.; Cai, W.-T.; Jin, H.-Z.; Yu, D.-H.; Sun, J.-H.; Yu, J. Effectiveness and Safety of Trabeculectomy along with Amniotic Membrane Transplantation on Glaucoma: A Systematic Review. J. Ophthalmol. 2020, 2020, 3949735. [Google Scholar] [CrossRef]
  51. Tym, W.H.; Seah, S.K. Augmentation of filtering blebs with perfluoropropane gas bubble: An experimental and pilot clinical study. Ophthalmology 1999, 106, 545–549. [Google Scholar] [CrossRef] [PubMed]
  52. Papaconstantinou, D.; Diagourtas, A.; Petrou, P.; Rouvas, A.; Vergados, A.; Koutsandrea, C.; Georgalas, I. Trabeculectomy with Healaflow versus Trabeculectomy for the Treatment of Glaucoma: A Case-Control Study. J. Ophthalmol. 2015, 2015, 836269. [Google Scholar] [CrossRef] [Green Version]
  53. Mudhol, R.; Bansal, R. Cross-linked hyaluronic acid viscoelastic scleral implant in trabeculectomy. Indian J. Ophthalmol. 2021, 69, 1135–1141. [Google Scholar] [CrossRef] [PubMed]
  54. Roy, S.; Thi, H.D.; Feusier, M.; Mermoud, A. Crosslinked Sodium Hyaluronate Implant in Deep Sclerectomy for the Surgical Treatment of Glaucoma. Eur. J. Ophthalmol. 2011, 22, 70–76. [Google Scholar] [CrossRef] [PubMed]
  55. Wu, L.; Liu, J.; Chang, X.; Zheng, Y. The therapeutic effect of Healaflow in glaucoma surgery. Am. J. Transl. Res. 2021, 13, 9729–9735. [Google Scholar]
  56. Wang, X.; Dai, W.-W.; Dang, Y.-L.; Hong, Y.; Zhang, C. Five Years’ Outcomes of Trabeculectomy with Cross-linked Sodium Hyaluronate Gel Implantation for Chinese Glaucoma Patients. Chin. Med. J. 2018, 131, 1562–1568. [Google Scholar] [CrossRef]
  57. Breusegem, C.; Spielberg, L.; Van Ginderdeuren, R.; Vandewalle, E.; Renier, C.; Van de Veire, S.; Fieuws, S.; Zeyen, T.; Stalmans, I. Preoperative Nonsteroidal Anti-inflammatory Drug or Steroid and Outcomes after Trabeculectomy: A Randomized Controlled Trial. Ophthalmology 2010, 117, 1324–1330. [Google Scholar] [CrossRef]
  58. Armstrong, J.J.; Welk, B.K.; Reid, J.N.; Kansal, V.; Hutnik, C.M. Secondary surgical intervention after primary glaucoma filtration surgery: An Ontario population-based study. Can. J. Ophthalmol. 2019, 54, 212–222. [Google Scholar] [CrossRef]
  59. Yamanaka, O.; Ikeda, K.; Saika, S.; Miyazaki, K.I.; Ooshima, A.; Ohnishi, Y. Gene transfer of Smad7 modulates injury-induced conjunctival wound healing in mice. Mol. Vis. 2006, 12, 841–851. [Google Scholar]
  60. Muppala, S.; Raghunathan, V.K.; Jalilian, I.; Thomasy, S.; Murphy, C.J. YAP and TAZ are distinct effectors of corneal myofibroblast transformation. Exp. Eye Res. 2019, 180, 102–109. [Google Scholar] [CrossRef]
  61. Seet, L.-F.; Su, R.; Toh, L.Z.; Wong, T.T. In vitro analyses of the anti-fibrotic effect of SPARC silencing in human Tenon’s fibroblasts: Comparisons with mitomycin C. J. Cell. Mol. Med. 2012, 16, 1245–1259. [Google Scholar] [CrossRef] [PubMed]
  62. Seet, L.-F.; Su, R.; Barathi, V.A.; Lee, W.S.; Poh, R.; Heng, Y.M.; Manser, E.; Vithana, E.N.; Aung, T.; Weaver, M.; et al. SPARC Deficiency Results in Improved Surgical Survival in a Novel Mouse Model of Glaucoma Filtration Surgery. PLoS ONE 2010, 5, e9415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lee, E.J.; Han, J.C.; Park, D.Y.; Cho, J.; Kee, C. Effect of connective tissue growth factor gene editing using adeno-associated virus–mediated CRISPR–Cas9 on rabbit glaucoma filtering surgery outcomes. Gene Ther. 2021, 28, 277–286. [Google Scholar] [CrossRef] [PubMed]
  64. Stahnke, T.; Kowtharapu, B.S.; Stachs, O.; Schmitz, K.-P.; Wurm, J.; Wree, A.; Guthoff, R.F.; Hovakimyan, M. Suppression of TGF-β pathway by pirfenidone decreases extracellular matrix deposition in ocular fibroblasts in vitro. PLoS ONE 2017, 12, e0172592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chun, Y.Y.; Yap, Z.L.; Seet, L.F.; Chan, H.H.; Toh, L.Z.; Chu, S.W.L.; Lee, Y.S.; Wong, T.T.; Tan, T.T.Y. Positive-charge tuned gelatin hydrogel-siSPARC injectable for siRNA anti-scarring therapy in post glaucoma filtration surgery. Sci. Rep. 2021, 11, 1–14. [Google Scholar] [CrossRef]
  66. Thijssen, V.L.; Paulis, Y.W.; Nowak-Sliwinska, P.; Deumelandt, K.L.; Hosaka, K.; Soetekouw, P.M.; Cimpean, A.M.; Raica, M.; Pauwels, P.; Oord, J.J.V.D.; et al. Targeting PDGF-mediated recruitment of pericytes blocks vascular mimicry and tumor growth. J. Pathol. 2018, 246, 447–458. [Google Scholar] [CrossRef] [Green Version]
  67. Lin, X.; Wen, J.; Liu, R.; Gao, W.; Qu, B.; Yu, M. Nintedanib inhibits TGF-β-induced myofibroblast transdifferentiation in human Tenon’s fibroblasts. Mol. Vis. 2018, 24, 789–800. [Google Scholar]
  68. Gehring, S.; Hoerauf, H.; Laqua, H.; Kirchner, H.; Klüter, H. Preparation of autologous platelets for the ophthalmologic treatment of macular holes. Transfusion 1999, 39, 144–148. [Google Scholar] [CrossRef]
  69. Adachi, K.; Asada, Y.; Hirakata, T.; Onoue, M.; Iwamoto, S.; Kasuga, T.; Matsuda, A. Alteration of gene expression in mice after glaucoma filtration surgery. Sci. Rep. 2020, 10, 15036. [Google Scholar] [CrossRef]
  70. Van Bergen, T.; Marshall, D.; Van De Veire, S.; Vandewalle, E.; Moons, L.; Herman, J.; Smith, V.; Stalmans, I. The Role of LOX and LOXL2 in Scar Formation After Glaucoma Surgery. Investig. Ophthalmol. Vis. Sci. 2013, 54, 5788–5796. [Google Scholar] [CrossRef]
  71. Sen, E.; Balikoglu-Yilmaz, M.; Bozdag-Pehlivan, S.; Sungu, N.; Aksakal, F.N.; Altinok, A.; Tuna, T.; Unlu, N.; Ustun, H.; Koklu, G.; et al. Effect of Doxycycline on Postoperative Scarring After Trabeculectomy in an Experimental Rabbit Model. J. Ocul. Pharmacol. Ther. 2010, 26, 399–406. [Google Scholar] [CrossRef]
  72. Chakraborti, S.; Mandal, M.; Das, S.; Mandal, A.; Chakraborti, T. Regulation of matrix metalloproteinases: An overview. Mol. Cell. Biochem. 2003, 253, 269–285. [Google Scholar] [CrossRef]
  73. Stillman, B. Histone Modifications: Insights into Their Influence on Gene Expression. Cell 2018, 175, 6–9. [Google Scholar] [CrossRef] [Green Version]
  74. Johnson, K.T.M.; Rödicker, F.; Heise, K.; Heinz, C.; Steuhl, K.-P.; Pützer, B.M.; Hudde, T. Adenoviral p53 gene transfer inhibits human Tenon’s capsule fibroblast proliferation. Br. J. Ophthalmol. 2005, 89, 508–512. [Google Scholar] [CrossRef]
  75. Perkins, T.W.; Faha, B.; Ni, M.; Kiland, J.A.; Poulsen, G.L.; Antelman, D.; Atencio, I.; Shinoda, J.; Sinha, D.; Brumback, L.; et al. Adenovirus-mediated gene therapy using human p21WAF-1/Cip-1 to prevent wound healing in a rabbit model of glaucoma filtration surgery. Arch. Ophthalmol. 2002, 120, 941–949. [Google Scholar] [CrossRef] [Green Version]
  76. Yang, J.-G.; Deng, Y.; Zhou, L.-X.; Li, X.-Y.; Sun, P.-R.; Sun, N.-X. Overexpression of CDKN1B Inhibits Fibroblast Proliferation in a Rabbit Model of Experimental Glaucoma Filtration Surgery. Investig. Ophthalmol. Vis. Sci. 2013, 54, 343–352. [Google Scholar] [CrossRef] [Green Version]
  77. Malhotra, C.; Jain, A.K. Human amniotic membrane transplantation: Different modalities of its use in ophthalmology. World J. Transplant. 2014, 4, 111–121. [Google Scholar] [CrossRef]
  78. Schultz, T.; Schojai, M.; Kersten-Gomez, I.; Matthias, E.; Boecker, J.; Dick, H.B. Ab externo device for the treatment of glaucoma: Direct flow from the anterior chamber to the ocular surface. J. Cataract. Refract. Surg. 2020, 46, 941–943. [Google Scholar] [CrossRef]
  79. Gressel, M.G.; Parrish, R.K.; Folberg, R. 5-Fluorouracil and Glaucoma Filtering Surgery. Ophthalmology 1984, 91, 378–383. [Google Scholar] [CrossRef]
  80. Smith, S.; D’Amore, P.; Dreyer, E.B. Comparative Toxicity of Mitomycin C and 5-Fluorouracil In Vitro. Am. J. Ophthalmol. 1994, 118, 332–337. [Google Scholar] [CrossRef]
  81. Jampel, H.D. Effect of Brief Exposure to Mitomycin C on Viability and Proliferation of Cultured Human Tenon’s Capsule Fibroblasts. Ophthalmology 1992, 99, 1471–1476. [Google Scholar] [CrossRef] [PubMed]
  82. Khaw, P.T.; Sherwood, M.B.; Doyle, J.W.; Smith, M.F.; Grierson, I.; McGorray, S.; Schultz, G.S. Intraoperative and post operative treatment with 5-Fluorouracil and mitomycin-c: Long term effects in vivo on subconjunctival and scleral fibroblasts. Int. Ophthalmol. 1992, 16, 381–385. [Google Scholar] [CrossRef] [PubMed]
  83. Khaw, P.T.; Ward, S.; Grierson, I.; Rice, N.S. Effect of beta radiation on proliferating human Tenon’s capsule fibroblasts. Br. J. Ophthalmol. 1991, 75, 580–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Constable, P.H.; Crowston, J.G.; Occleston, N.L.; Cordeiro, M.F.; Khaw, P.T. Long term growth arrest of human Tenon’s fibroblasts following single applications of beta radiation. Br. J. Ophthalmol. 1998, 82, 448–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Bedinger, D.; Lao, L.; Khan, S.; Lee, S.; Takeuchi, T.; Mirza, A.M. Development and characterization of human monoclonal antibodies that neutralize multiple TGFβ isoforms. mAbs 2015, 8, 389–404. [Google Scholar] [CrossRef] [Green Version]
  86. Akhurst, R.J.; Hata, A. Targeting the TGFβ signalling pathway in disease. Nat. Rev. Drug Discov. 2012, 11, 790–811. [Google Scholar] [CrossRef] [Green Version]
  87. Zhong, H.; Sun, G.; Lin, X.; Wu, K.; Yu, M. Evaluation of Pirfenidone as a New Postoperative Antiscarring Agent in Experimental Glaucoma Surgery. Investig. Ophthalmol. Vis. Sci. 2011, 52, 3136–3142. [Google Scholar] [CrossRef] [Green Version]
  88. Futakuchi, A.; Inoue, T.; Wei, F.-Y.; Inoue-Mochita, M.; Fujimoto, T.; Tomizawa, K.; Tanihara, H. YAP/TAZ Are Essential for TGF-β2–Mediated Conjunctival Fibrosis. Investig. Ophthalmol. Vis. Sci. 2018, 59, 3069–3078. [Google Scholar] [CrossRef] [Green Version]
  89. Cheng, J.-W.; Cheng, S.-W.; Wei, R.-L.; Lu, G.-C. Anti-vascular endothelial growth factor for control of wound healing in glaucoma surgery. Cochrane Database Syst. Rev. 2016, 2016, CD009782. [Google Scholar] [CrossRef]
  90. Wrana, J.L.; Kubota, T.; Zhang, Q.; Overall, C.M.; E Aubin, J.; Butler, W.T.; Sodek, J. Regulation of transformation-sensitive secreted phosphoprotein (SPPI/osteopontin) expression by transforming growth factor-β. Comparisons with expression of SPARC (secreted acidic cysteine-rich protein). Biochem. J. 1991, 273, 523–531. [Google Scholar] [CrossRef]
  91. Tripathi, R.; Sinha, N.R.; Kempuraj, D.; Balne, P.K.; Landreneau, J.R.; Juneja, A.; Webel, A.D.; Mohan, R.R. Evaluation of CRISPR/Cas9 mediated TGIF gene editing to inhibit corneal fibrosis in vitro. Exp. Eye Res. 2022, 220, 109113. [Google Scholar] [CrossRef] [PubMed]
  92. Sharma, A.; Sinha, N.R.; Siddiqui, S.; Mohan, R.R. Role of 5′TG3′-interacting factors (TGIFs) in Vorinostat (HDAC inhibitor)-mediated Corneal Fibrosis Inhibition. Mol. Vis. 2015, 21, 974–984. [Google Scholar] [PubMed]
  93. Tandon, A.; Tovey, J.C.; Waggoner, M.R.; Sharma, A.; Cowden, J.W.; Gibson, D.; Liu, Y.; Schultz, G.S.; Mohan, R.R. Vorinostat: A Potent Agent to Prevent and Treat Laser-induced Corneal Haze. J. Refract. Surg. 2012, 28, 285–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kitano, A.; Okada, Y.; Yamanka, O.; Shirai, K.; Mohan, R.R.; Saika, S. Therapeutic potential of Trichostatin A to control inflammatory and fibrogenic disorders of the ocular surface. Mol. Vis. 2010, 16, 2964–2973. [Google Scholar]
  95. Zhang, X.; Zhang, J.; Liu, Y.; Zhu, D.; Chen, D.; Zhang, Z.; Sun, Y. Pirfenidone inhibits fibroblast proliferation, migration or adhesion and reduces epidural fibrosis in rats via the PI3K/AKT signaling pathway. Biochem. Biophys. Res. Commun. 2021, 547, 183–191. [Google Scholar] [CrossRef]
  96. Jin, J.; Togo, S.; Kadoya, K.; Tulafu, M.; Namba, Y.; Iwai, M.; Watanabe, J.; Nagahama, K.; Okabe, T.; Hidayat, M.; et al. Pirfenidone attenuates lung fibrotic fibroblast responses to transforming growth factor-β1. Respir. Res. 2019, 20, 119. [Google Scholar] [CrossRef] [Green Version]
  97. Liu, X.; Yang, Y.; Guo, X.; Liu, L.; Wu, K.; Yu, M. The Antiangiogenesis Effect of Pirfenidone in Wound Healing In Vitro. J. Ocul. Pharmacol. Ther. 2017, 33, 693–703. [Google Scholar] [CrossRef]
  98. Samaeili, A.; Rahmani, S.; Hassanpour, K.; Meshksar, A.; Ansari, I.; Afsar-Aski, S.; Einollahi, B.; Pakravan, M. A new glaucoma drainage implant with the use of Polytetrafluoroethylene (PTFE). A pilot study. Rom. J. Ophthalmol. 2021, 65, 150–156. [Google Scholar] [CrossRef]
  99. White, T.C.; Berny, M.A.; Robinson, D.K.; Yin, H.; DeGrado, W.F.; Hanson, S.R.; McCarty, O.J.T. The leech product saratin is a potent inhibitor of platelet integrin α2β1 and von Willebrand factor binding to collagen. FEBS J. 2007, 274, 1481–1491. [Google Scholar] [CrossRef]
  100. McCluskey, P.; Molteno, A.; Wakefield, D.; Di Girolamo, N. Otago Glaucoma Surgery Outcome Study: The Pattern of Expression of MMPs and TIMPs in Bleb Capsules Surrounding Molteno Implants. Investig. Ophthalmol. Vis. Sci. 2009, 50, 2161–2164. [Google Scholar] [CrossRef] [Green Version]
  101. Tagalakis, A.D.; Madaan, S.; Larsen, S.D.; Neubig, R.R.; Khaw, P.T.; Rodrigues, I.; Yu-Wai-Man, C. In vitro and in vivo delivery of a sustained release nanocarrier-based formulation of an MRTF/SRF inhibitor in conjunctival fibrosis. J. Nanobiotechnol. 2018, 16, 1–11. [Google Scholar] [CrossRef] [Green Version]
  102. Hafner, A.; Bulyk, M.L.; Jambhekar, A.; Lahav, G. The multiple mechanisms that regulate p53 activity and cell fate. Nat. Rev. Mol. Cell Biol. 2019, 20, 199–210. [Google Scholar] [CrossRef]
  103. Li, N.; Zhang, R. Silencing of p53 reduces cell migration in human Tenon’s fibroblasts induced by TGF-β. Int. Ophthalmol. 2020, 40, 1509–1516. [Google Scholar] [CrossRef]
  104. Seibold, L.K.; Sherwood, M.B.; Kahook, M.Y. Wound Modulation After Filtration Surgery. Surv. Ophthalmol. 2012, 57, 530–550. [Google Scholar] [CrossRef]
  105. Bhagat, R.; Zhang, J.; Farooq, S.; Li, X.-Y. Comparison of the Release Profile and Pharmacokinetics of Intact and Fragmented Dexamethasone Intravitreal Implants in Rabbit Eyes. J. Ocul. Pharmacol. Ther. 2014, 30, 854–858. [Google Scholar] [CrossRef]
  106. Salmon, J. Chapter 13: Gonioscopy. Section 3: Diagnosis of glaucoma. In Glaucoma: Medical Diagnosis and Therapy; Saunders Ltd.: London, UK, 2009; p. 668. [Google Scholar]
  107. Lo, K.-J.; Ko, Y.-C.; Hwang, D.-K.; Liu, C.J.-L. The influence of topical non-steroidal anti-inflammatory drugs on the intraocular pressure lowering effect of topical prostaglandin analogues—A systemic review and meta-analysis. PLoS ONE 2020, 15, e0239233. [Google Scholar] [CrossRef]
  108. Almatlouh, A.; Bach-Holm, D.; Kessel, L. Steroids and nonsteroidal anti-inflammatory drugs in the postoperative regime after trabeculectomy—Which provides the better outcome? A systematic review and meta-analysis. Acta Ophthalmol. 2018, 97, 146–157. [Google Scholar] [CrossRef]
  109. Yuen, D.; Buys, Y.; Jin, Y.-P.; Alasbali, T.; Smith, M.; Trope, G.E. Corticosteroids Versus NSAIDs on Intraocular Pressure and the Hypertensive Phase After Ahmed Glaucoma Valve Surgery. Eur. J. Gastroenterol. Hepatol. 2011, 20, 439–444. [Google Scholar] [CrossRef]
  110. Shapiro, M.S.; A Thoft, R.; Friend, J.; Parrish, R.K.; Gressel, M.G. 5-Fluorouracil toxicity to the ocular surface epithelium. Investig. Ophthalmol. Vis. Sci. 1985, 26, 580–583. [Google Scholar]
  111. DeBry, P.W.; Perkins, T.W.; Heatley, G.; Kaufman, P.; Brumback, L.C. Incidence of Late-Onset Bleb-Related Complications Following Trabeculectomy With Mitomycin. Arch. Ophthalmol. 2002, 120, 297–300. [Google Scholar] [CrossRef] [Green Version]
  112. Muckley, E.D.; Lehrer, R.A. Late-Onset Blebitis/Endophthalmitis: Incidence and Outcomes with Mitomycin C. Optom. Vis. Sci. 2004, 81, 499–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Bindlish, R.; Condon, G.P.; Schlosser, J.D.; D’Antonio, J.; Lauer, K.B.; Lehrer, R. Efficacy and safety of mitomycin-C in primary trabeculectomy: Five-year follow-up. Ophthalmology 2002, 109, 1336–1341. [Google Scholar] [CrossRef] [PubMed]
  114. Anand, N.; Arora, S.; Clowes, M. Mitomycin C augmented glaucoma surgery: Evolution of filtering bleb avascularity, transconjunctival oozing, and leaks. Br. J. Ophthalmol. 2006, 90, 175–180. [Google Scholar] [CrossRef] [PubMed]
  115. Wolters, J.E.; van Mechelen, R.J.; Al Majidi, R.; Pinchuk, L.; Webers, C.A.; Beckers, H.J.; Gorgels, T.G. History, presence, and future of mitomycin C in glaucoma filtration surgery. Curr. Opin. Ophthalmol. 2021, 32, 148–159. [Google Scholar] [CrossRef] [PubMed]
  116. Grover, D.S.; Kornmann, H.L.; Fellman, R.L. Historical Considerations and Innovations in the Perioperative Use of Mitomycin C for Glaucoma Filtration Surgery and Bleb Revisions. Eur. J. Gastroenterol. Hepatol. 2020, 29, 226–235. [Google Scholar] [CrossRef]
  117. Khouri, A.S.; Huang, G.; Huang, L.Y. Intraoperative Injection vs Sponge-applied Mitomycin C during Trabeculectomy: A One-year Study. Curr. J. Glaucoma Pract. DVD 2017, 11, 101–106. [Google Scholar] [CrossRef]
  118. Do, J.L.; Xu, B.Y.; Wong, B.; Camp, A.; Ngai, P.; Long, C.; Proudfoot, J.; Moghimi, S.; Yan, D.; Welsbie, D.S.; et al. A Randomized Controlled Trial Comparing Subconjunctival Injection to Direct Scleral Application of Mitomycin C in Trabeculectomy. Am. J. Ophthalmol. 2020, 220, 45–52. [Google Scholar] [CrossRef]
  119. Palanca-Capistrano, A.M.; Hall, J.; Cantor, L.B.; Morgan, L.; Hoop, J.; WuDunn, D. Long-term Outcomes of Intraoperative 5-Fluorouracil versus Intraoperative Mitomycin C in Primary Trabeculectomy Surgery. Ophthalmology 2009, 116, 185–190. [Google Scholar] [CrossRef]
  120. Kim, T.-I.; Tchah, H.; Lee, S.-A.; Sung, K.; Cho, B.J.; Kook, M.S. Apoptosis in Keratocytes Caused by Mitomycin C. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1912–1917. [Google Scholar] [CrossRef] [Green Version]
  121. Yap, Z.L.; Seet, L.-F.; Chu, S.W.; Toh, L.Z.; Ibrahim, F.I.; Wong, T.T. Effect of valproic acid on functional bleb morphology in a rabbit model of minimally invasive surgery. Br. J. Ophthalmol. 2022, 106, 1028–1036. [Google Scholar] [CrossRef]
  122. Seet, L.-F.; Yap, Z.L.; Chu, S.W.L.; Toh, L.Z.; Ibrahim, F.I.; Teng, X.; Wong, T.T. Effects of Valproic Acid and Mitomycin C Combination Therapy in a Rabbit Model of Minimally Invasive Glaucoma Surgery. Transl. Vis. Sci. Technol. 2022, 11, 30. [Google Scholar] [CrossRef]
  123. Cook, C.; Perrott, A.; Mustak, H.; Courtright, P.; Wetter, J.; du Toit, N. Randomised clinical trial of trabeculectomy with mitomycin-C versus trabeculectomy with beta radiation. S. Afr. Ophthalmol. J. 2018, 13, 11–14. [Google Scholar] [CrossRef]
  124. CAT-152 0102 Trabeculectomy Study Group. A Phase III Study of Subconjunctival Human Anti–Transforming Growth Factor β2 Monoclonal Antibody (CAT-152) to Prevent Scarring after First-Time Trabeculectomy. Ophthalmology 2007, 114, 1822–1830.e2. [Google Scholar] [CrossRef]
  125. Saika, S.; Ikeda, K.; Yamanaka, O.; Miyamoto, T.; Ohnishi, Y.; Sato, M.; Muragaki, Y.; Ooshima, A.; Nakajima, Y.; Kao, W.W.-Y.; et al. Expression of Smad7 in Mouse Eyes Accelerates Healing of Corneal Tissue after Exposure to Alkali. Am. J. Pathol. 2005, 166, 1405–1418. [Google Scholar] [CrossRef] [Green Version]
  126. Gupta, S.; Rodier, J.T.; Sharma, A.; Giuliano, E.A.; Sinha, P.R.; Hesemann, N.P.; Ghosh, A.; Mohan, R.R. Targeted AAV5-Smad7 gene therapy inhibits corneal scarring in vivo. PLoS ONE 2017, 12, e0172928. [Google Scholar] [CrossRef] [Green Version]
  127. Myrna, K.E.; Mendonsa, R.; Russell, P.; Pot, S.A.; Liliensiek, S.J.; Jester, J.V.; Nealey, P.F.; Brown, D.; Murphy, C.J. Substratum Topography Modulates Corneal Fibroblast to Myofibroblast Transformation. Investig. Ophthalmol. Vis. Sci. 2012, 53, 811–816. [Google Scholar] [CrossRef] [Green Version]
  128. Lim, R.R.; Tan, A.; Liu, Y.-C.; Barathi, V.A.; Mohan, R.R.; Mehta, J.S.; Chaurasia, S.S. ITF2357 transactivates Id3 and regulate TGFβ/BMP7 signaling pathways to attenuate corneal fibrosis. Sci. Rep. 2016, 6, 20841. [Google Scholar] [CrossRef] [Green Version]
  129. Sung, M.S.; Eom, G.H.; Kim, S.J.; Kim, S.Y.; Heo, H.; Park, S.W. Trichostatin A Ameliorates Conjunctival Fibrosis in a Rat Trabeculectomy Model. Investig. Ophthalmol. Vis. Sci. 2018, 59, 3115–3123. [Google Scholar] [CrossRef] [Green Version]
  130. Pan, H.-W.; Xu, J.-T.; Chen, J.-S. Pioglitazone inhibits TGFβ induced keratocyte transformation to myofibroblast and extracellular matrix production. Mol. Biol. Rep. 2011, 38, 4501–4508. [Google Scholar] [CrossRef]
  131. Saika, S.; Yamanaka, O.; Okada, Y.; Miyamoto, T.; Kitano, A.; Flanders, K.C.; Ohnishi, Y.; Nakajima, Y.; Kao, W.W.-Y.; Ikeda, K. Effect of overexpression of pparγ on the healing process of corneal alkali burn in mice. Am. J. Physiol. Physiol. 2007, 293, C75–C86. [Google Scholar] [CrossRef]
  132. Zhang, F.; Liu, K.; Cao, M.; Qu, J.; Zhou, D.; Pan, Z.; Duan, X.; Zhou, Y. Rosiglitazone Treatment Prevents Postoperative Fibrosis in a Rabbit Model of Glaucoma Filtration Surgery. Investig. Ophthalmol. Vis. Sci. 2019, 60, 2743–2752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Meyer-Ter-Vehn, T.; Gebhardt, S.; Sebald, W.; Buttmann, M.; Grehn, F.; Schlunck, G.; Knaus, P. p38 Inhibitors Prevent TGF-β–Induced Myofibroblast Transdifferentiation in Human Tenon Fibroblasts. Investig. Ophthalmol. Vis. Sci. 2006, 47, 1500–1509. [Google Scholar] [CrossRef]
  134. Liang, M.; Lv, J.; Chu, H.; Wang, J.; Chen, X.; Zhu, X.; Xue, Y.; Guan, M.; Zou, H. Vertical inhibition of PI3K/Akt/mTOR signaling demonstrates in vitro and in vivo anti-fibrotic activity. J. Dermatol. Sci. 2014, 76, 104–111. [Google Scholar] [CrossRef] [PubMed]
  135. Kang, X.; Shen, Y.; Zhao, H.; Wang, Z.; Guan, W.; Ge, R.; Wang, R.; Tai, X. Anti-scarring effect of rapamycin in rabbits following glaucoma filtering surgery. Nan Fang Yi Ke Da Xue Xue Bao J. South. Med. Univ. 2018, 38, 1389–1394. [Google Scholar] [CrossRef]
  136. Pitha, I.; Oglesby, E.; Chow, A.; Kimball, E.; Pease, M.E.; Schaub, J.; Quigley, H. Rho-Kinase Inhibition Reduces Myofibroblast Differentiation and Proliferation of Scleral Fibroblasts Induced by Transforming Growth Factor β and Experimental Glaucoma. Transl. Vis. Sci. Technol. 2018, 7, 6. [Google Scholar] [CrossRef] [Green Version]
  137. Kaibuchi, K.; Kuroda, S.; Amano, M. Regulation of the Cytoskeleton and Cell Adhesion by the Rho Family GTPases in Mammalian Cells. Annu. Rev. Biochem. 1999, 68, 459–486. [Google Scholar] [CrossRef] [Green Version]
  138. Honjo, M.; Tanihara, H.; Kameda, T.; Kawaji, T.; Yoshimura, N.; Araie, M. Potential Role of Rho-Associated Protein Kinase Inhibitor Y-27632 in Glaucoma Filtration Surgery. Investig. Ophthalmol. Vis. Sci. 2007, 48, 5549–5557. [Google Scholar] [CrossRef] [Green Version]
  139. Tandon, A.; Tovey, J.C.K.; Sharma, A.; Gupta, R.; Mohan, R.R. Role of Transforming Growth Factor Beta in Corneal Function, Biology and Pathology. Curr. Mol. Med. 2010, 10, 565–578. [Google Scholar] [CrossRef]
  140. Mohan, R.R.; Sinha, S.; Tandon, A.; Gupta, R.; Tovey, J.C.K.; Sharma, A. Efficacious and Safe Tissue-Selective Controlled Gene Therapy Approaches for the Cornea. PLoS ONE 2011, 6, e18771. [Google Scholar] [CrossRef]
  141. Anumanthan, G.; Sharma, A.; Waggoner, M.; Hamm, C.W.; Gupta, S.; Hesemann, N.P.; Mohan, R.R. Efficacy and Safety Comparison Between Suberoylanilide Hydroxamic Acid and Mitomycin C in Reducing the Risk of Corneal Haze After PRK Treatment In Vivo. J. Refract. Surg. 2017, 33, 834–839. [Google Scholar] [CrossRef]
  142. Pitre, T.; Mah, J.; Helmeczi, W.; Khalid, M.F.; Cui, S.; Zhang, M.; Husnudinov, R.; Su, J.; Banfield, L.; Guy, B.; et al. Medical treatments for idiopathic pulmonary fibrosis: A systematic review and network meta-analysis. Thorax 2022, 77, 1243–1250. [Google Scholar] [CrossRef] [PubMed]
  143. Ruwanpura, S.M.; Thomas, B.J.; Bardin, P.G. Pirfenidone: Molecular Mechanisms and Potential Clinical Applications in Lung Disease. Am. J. Respir. Cell Mol. Biol. 2020, 62, 413–422. [Google Scholar] [CrossRef] [PubMed]
  144. Lin, X.; Yu, M.; Wu, K.; Yuan, H.; Zhong, H. Effects of Pirfenidone on Proliferation, Migration, and Collagen Contraction of Human Tenon’s Fibroblasts In Vitro. Investig. Ophthalmol. Vis. Sci. 2009, 50, 3763–3770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Hwang, Y.H.; Jung, S.-A.; Lyu, J.; Kim, Y.Y.; Lee, J.H. Transforming Growth Factor-β1–induced Human Subconjunctival Fibrosis is Mediated by MicroRNA 143/145 Expression. Investig. Ophthalmol. Vis. Sci. 2019, 60, 2064–2071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Kang, M.H.; Oh, D.-J.; Kang, J.-H.; Rhee, D.J. Regulation of SPARC by Transforming Growth Factor β2 in Human Trabecular Meshwork. Investig. Ophthalmol. Vis. Sci. 2013, 54, 2523–2532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Seet, L.F.; Tan, Y.F.; Toh, L.Z.; Chu, S.W.; Lee, Y.S.; Venkatraman, S.S.; Wong, T.T. Targeted therapy for the post-operative conjunctiva: SPARC silencing reduces collagen deposition. Br. J. Ophthalmol. 2018, 102, 1460–1470. [Google Scholar] [CrossRef] [PubMed]
  148. Heldin, C.-H.; Lennartsson, J.; Westermark, B. Involvement of platelet-derived growth factor ligands and receptors in tumorigenesis. J. Intern. Med. 2018, 283, 16–44. [Google Scholar] [CrossRef] [Green Version]
  149. Akiyama, H.; Kachi, S.; E Silva, R.L.; Umeda, N.; Hackett, S.F.; McCauley, D.; McCauley, T.; Zoltoski, A.; Epstein, D.M.; Campochiaro, P.A. Intraocular injection of an aptamer that binds PDGF-B: A potential treatment for proliferative retinopathies. J. Cell. Physiol. 2006, 207, 407–412. [Google Scholar] [CrossRef]
  150. Li, Z.; Van Bergen, T.; Van De Veire, S.; Van De Vel, I.; Moreau, H.; Dewerchin, M.; Maudgal, P.C.; Zeyen, T.; Spileers, W.; Moons, L.; et al. Inhibition of Vascular Endothelial Growth Factor Reduces Scar Formation after Glaucoma Filtration Surgery. Investig. Ophthalmol. Vis. Sci. 2009, 50, 5217–5225. [Google Scholar] [CrossRef]
  151. Sengupta, S.; Venkatesh, R.; Ravindran, R.D. Safety and Efficacy of Using Off-label Bevacizumab Versus Mitomycin C to Prevent Bleb Failure in a Single-site Phacotrabeculectomy by a Randomized Controlled Clinical Trial. Eur. J. Gastroenterol. Hepatol. 2012, 21, 450–459. [Google Scholar] [CrossRef]
  152. Kaushik, J.; Parihar, J.K.S.; Jain, V.K.; Gupta, S.; Nath, P.; Durgapal, P.; Ram, J. Efficacy of Bevacizumab Compared to Mitomycin C Modulated Trabeculectomy in Primary Open Angle Glaucoma: A One-Year Prospective Randomized Controlled Study. Curr. Eye Res. 2017, 42, 217–224. [Google Scholar] [CrossRef]
  153. Kopsinis, G.; Tsoukanas, D.; Kopsini, D.; Filippopoulos, T. Intracameral Bevacizumab Versus Sub-Tenon’s Mitomycin C as Adjuncts to Trabeculectomy: 3-Year Results of a Prospective Randomized Study. J. Clin. Med. 2021, 10, 2054. [Google Scholar] [CrossRef]
  154. Zuo, L.; Zhang, J.; Xu, X. Combined Application of Bevacizumab and Mitomycin C or Bevacizumab and 5-Fluorouracil in Experimental Glaucoma Filtration Surgery. J. Ophthalmol. 2018, 2018, 8965709. [Google Scholar] [CrossRef]
  155. Zhang, Y.; Zhu, S.; Xu, X.; Zuo, L. In Vitro Study of Combined Application of Bevacizumab and 5-Fluorouracil or Bevacizumab and Mitomycin C to Inhibit Scar Formation in Glaucoma Filtration Surgery. J. Ophthalmol. 2019, 2019, 7419571. [Google Scholar] [CrossRef] [Green Version]
  156. Kahook, M.Y. Bleb Morphology and Vascularity after Trabeculectomy with Intravitreal Ranibizumab: A Pilot Study. Am. J. Ophthalmol. 2010, 150, 399–403.e1. [Google Scholar] [CrossRef]
  157. Zalta, A.H. Intracameral Tissue Plasminogen Activator Use in a Large Series of Eyes with Valved Glaucoma Drainage Implants. Arch. Ophthalmol. 2002, 120, 1487–1493. [Google Scholar] [CrossRef] [Green Version]
  158. Barequet, D.; Rosenfeld, E.; Rabina, G.; Shemesh, G.; Kurtz, S. Intracameral tissue plasminogen activator in trabeculectomy: A 1-year prospective, randomized, controlled study. Int. Ophthalmol. 2020, 40, 1641–1646. [Google Scholar] [CrossRef]
  159. WuDunn, D. Intracameral Urokinase for Dissolution of Fibrin or Blood Clots After Glaucoma Surgery. Am. J. Ophthalmol. 1997, 124, 693–695. [Google Scholar] [CrossRef]
  160. Min, J.; Lukowski, Z.L.; Levine, M.A.; Meyers, C.A.; Beattie, A.R.; Schultz, G.S.; Samuelson, D.A.; Sherwood, M.B. Prevention of Ocular Scarring Post Glaucoma Filtration Surgery Using the Inflammatory Cell and Platelet Binding Modulator Saratin in a Rabbit Model. PLoS ONE 2012, 7, e35627. [Google Scholar] [CrossRef]
  161. Vallet, S.D.; Ricard-Blum, S. Lysyl oxidases: From enzyme activity to extracellular matrix cross-links. Essays Biochem. 2019, 63, 349–364. [Google Scholar] [CrossRef]
  162. Chen, W.; Yang, A.; Jia, J.; Popov, Y.V.; Schuppan, D.; You, H. Lysyl Oxidase (LOX) Family Members: Rationale and Their Potential as Therapeutic Targets for Liver Fibrosis. Hepatology 2020, 72, 729–741. [Google Scholar] [CrossRef] [PubMed]
  163. Rodríguez, C.; Martínez-González, J. The Role of Lysyl Oxidase Enzymes in Cardiac Function and Remodeling. Cells 2019, 8, 1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Park, H.-Y.L.; Kim, J.H.; Park, C.K. Lysyl Oxidase-Like 2 Level and Glaucoma Surgical Outcomes. Investig. Ophthalmol. Vis. Sci. 2014, 55, 3337–3343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Wong, T.T.; Sethi, C.; Daniels, J.T.; Limb, G.; Murphy, G.; Khaw, P.T. Matrix Metalloproteinases in Disease and Repair Processes in the Anterior Segment. Surv. Ophthalmol. 2002, 47, 239–256. [Google Scholar] [CrossRef] [PubMed]
  166. Daniels, J.T.; Cambrey, A.D.; Occleston, N.L.; Garrett, Q.; Tarnuzzer, R.; Schultz, G.S.; Khaw, P.T. Matrix Metalloproteinase Inhibition Modulates Fibroblast-Mediated Matrix Contraction and Collagen Production In Vitro. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1104–1110. [Google Scholar] [CrossRef]
  167. Suh, W.; Han, K.E.; Han, J.R. Safety of Using Matrix Metalloproteinase Inhibitor in Experimental Glaucoma Filtration Surgery. J. Korean Med. Sci. 2017, 32, 666–671. [Google Scholar] [CrossRef]
  168. Mohamed-Ahmed, A.H.A.; Lockwood, A.; Li, H.; Bailly, M.; Khaw, P.T.; Brocchini, S. An Ilomastat-CD Eye Drop Formulation to Treat Ocular Scarring. Investig. Ophthalmol. Vis. Sci. 2017, 58, 3425–3431. [Google Scholar] [CrossRef] [Green Version]
  169. Fernando, O.; Tagalakis, A.D.; Awwad, S.; Brocchini, S.; Khaw, P.T.; Hart, S.L.; Yu-Wai-Man, C. Development of Targeted siRNA Nanocomplexes to Prevent Fibrosis in Experimental Glaucoma Filtration Surgery. Mol. Ther. 2018, 26, 2812–2822. [Google Scholar] [CrossRef] [Green Version]
  170. Yu-Wai-Man, C.; Tagalakis, A.D.; Manunta, M.D.; Hart, S.L.; Khaw, P.T. Receptor-targeted liposome-peptide-siRNA nanoparticles represent an efficient delivery system for MRTF silencing in conjunctival fibrosis. Sci. Rep. 2016, 6, 21881. [Google Scholar] [CrossRef]
  171. Sharma, A.; Anumanthan, G.; Reyes, M.; Chen, H.; Brubaker, J.W.; Siddiqui, S.; Gupta, S.; Rieger, F.G.; Mohan, R.R. Epigenetic Modification Prevents Excessive Wound Healing and Scar Formation After Glaucoma Filtration Surgery. Investig. Ophthalmol. Vis. Sci. 2016, 57, 3381–3389. [Google Scholar] [CrossRef] [Green Version]
  172. Rodgers, C.D.; Sherwood, M.B.; Lukowski, Z.L.; Min, J.; Martorana, G.M.; Wilson, M.-K.; Schaefer, J.L.; A Levine, M.; A Meyers, C.; Blake, C.R.; et al. Modulating Ocular Scarring in Glaucoma Filtration Surgery Using the Epigenetic Adjunct Suberoylanilide Hydroxamic Acid. Curr. J. Glaucoma Pract. DVD 2019, 13, 37–41. [Google Scholar] [CrossRef]
  173. Yan, W.; Liao, R.; Li, N. Expression of p53 and its effect on microRNA-29b expression in the process of normal human conjunctival fibroblasts transforming to myofibroblast. Zhonghua Shiyan Yanke Zazhi/Chin. J. Exp. Ophthalmol. 2018, 36, 676–681. [Google Scholar] [CrossRef]
  174. Park, K.H.; Kim, D.M.; Youn, D.H. Topical cyclosporine and glaucoma drainage implant surgery in rabbits. Ophthalmic Surg. Lasers 1996, 27, 452–458. [Google Scholar] [CrossRef]
  175. Zhigalskiaya, T.A.; Dzyman, A.N.; Krylova, A.A.; Krivosheina, O.I.; Khlusov, I.A. Patterns of conjunctival and scleral regeneration after intraoperative application of cyclosporin A solution in rabbits with steroid-induced glaucoma. Bull. Sib. Med. 2021, 20, 36–43. [Google Scholar] [CrossRef]
  176. Dai, Z.-X.; Song, X.-L.; Yu, X.-B.; Sun, J.-G.; Sun, X.-H. Cyclosporine A-loaded drug delivery systems inhibit scar formation after glaucoma surgery in rabbits. Chin. Med. J. 2019, 132, 1381–1384. [Google Scholar] [CrossRef]
  177. A Lattanzio, F.; Crouch, E.R.; Mitrev, P.V.; Williams, P.B.; Allen, R.C. Cyclosporin as an Adjunct to Glaucoma Filtration Surgery. Eur. J. Gastroenterol. Hepatol. 2005, 14, 441–447. [Google Scholar] [CrossRef]
  178. Fakhraie, G.; Lopes, J.F.; Spaeth, G.L.; Almodin, J.; Ichhpujani, P.; Moster, M.R. Effects of postoperative cyclosporine ophthalmic emulsion 0.05% (Restasis) following glaucoma surgery. Clin. Exp. Ophthalmol. 2009, 37, 842–848. [Google Scholar] [CrossRef]
  179. Yan, Z.-C.; Bai, Y.-J.; Tian, Z.; Hu, H.-Y.; You, X.-H.; Lin, J.-X.; Liu, S.-R.; Zhuo, Y.-H.; Luo, R.-J. Anti-proliferation effects of Sirolimus sustained delivery film in rabbit glaucoma filtration surgery. Mol. Vis. 2011, 17, 2495–2506. [Google Scholar]
  180. Barton, K.; Budenz, D.L.; Khaw, P.T.; Tseng, S.C. Glaucoma filtration surgery using amniotic membrane transplantation. Investig. Ophthalmol. Vis. Sci. 2001, 42, 1762–1768. [Google Scholar]
  181. Roque, J.; Vaz, F.T.; Basto, R.; Henriques, S.; Lopes, A.S.; Silva, D.; Santos, J.; Pires, G.; Lisboa, M.; Prieto, I. Use of Amniotic Membrane in MMC-Augmented Trabeculectomy: A Retrospective Comparative Study. Clin. Ophthalmol. 2021, ume 15, 4527–4533. [Google Scholar] [CrossRef]
  182. Wang, X.; Khan, R.; Coleman, A. Device-modified trabeculectomy for glaucoma. Cochrane Database Syst. Rev. 2015, 2015, CD010472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Elwehidy, A.S.; Abouelkheir, H.Y.; Samra, W.A.; Wagdy, F.M.; Abdelkader, A.M.E. Trabeculectomy with Ologen implant versus perfluoropropane gas bubble for open angle glaucoma in pseudophakic eyes. Int. J. Ophthalmol. 2021, 14, 510–516. [Google Scholar] [CrossRef] [PubMed]
  184. Lu, L.J.; Hall, L.; Liu, J. Improving Glaucoma Surgical Outcomes with Adjunct Tools. J. Curr. Glaucoma Pract. 2018, 12, 19–28. [Google Scholar] [CrossRef] [PubMed]
  185. Harizman, N.; Du, J.; Tai, T.Y.T. A Prospective Randomized Trial of Ahmed Glaucoma Drainage Device Implantation with or without Ologen Collagen Matrix. Ophthalmol. Glaucoma 2020, 4, 421–426. [Google Scholar] [CrossRef]
  186. Fargione, R.A.; Tansuebchueasai, N.; Lee, R.; Tai, T.Y.T. Etiology and management of the hypertensive phase in glaucoma drainage-device surgery. Surv. Ophthalmol. 2018, 64, 217–224. [Google Scholar] [CrossRef]
  187. Özalp, O.; Ilgüy, S.; Atalay, E.; Şimşek, T.; Yıldırım, N. Risk factors for hypertensive phase after Ahmed glaucoma valve implantation. Int. Ophthalmol. 2021, 42, 147–156. [Google Scholar] [CrossRef]
  188. Bora, M.; Mundargi, R.C.; Chee, Y.; Wong, T.T.; Venkatraman, S.S. 5-Flurouracil microencapsulation and impregnation in hyaluronic acid hydrogel as composite drug delivery system for ocular fibrosis. Cogent Med. 2016, 3, 1182108. [Google Scholar] [CrossRef]
  189. Correa, S.; Grosskopf, A.K.; Hernandez, H.L.; Chan, D.; Yu, A.C.; Stapleton, L.M.; Appel, E.A. Translational Applications of Hydrogels. Chem. Rev. 2021, 121, 11385–11457. [Google Scholar] [CrossRef]
  190. Liu, W.; Borrell, M.A.; Venerus, D.C.; Mieler, W.F.; Kang-Mieler, J.J. Characterization of Biodegradable Microsphere-Hydrogel Ocular Drug Delivery System for Controlled and Extended Release of Ranibizumab. Transl. Vis. Sci. Technol. 2019, 8, 12. [Google Scholar] [CrossRef]
  191. Rudeen, K.M.; Liu, W.; Mieler, W.F.; Kang-Mieler, J.J. Simultaneous Release of Aflibercept and Dexamethasone from an Ocular Drug Delivery System. Curr. Eye Res. 2022, 47, 1034–1042. [Google Scholar] [CrossRef]
  192. Vandervoort, J.; Ludwig, A. Ocular drug delivery: Nanomedicine applications. Nanomedicine 2007, 2, 11–21. [Google Scholar] [CrossRef]
  193. Tilleul, P.; Denis, P.; Maignen, F.; Eléna, P.P.; Nordmann, J.P.; Leverge, R.; Rostene, W. Effects of Different Formulations of Mitoxantrone (Solutions, Nanospheres, Liposomes) on Glaucoma Surgery in Rabbits. Ophthalmic Res. 1997, 29, 218–226. [Google Scholar] [CrossRef]
  194. Simmons, S.T.; Sherwood, M.B.; A Nichols, D.; Penne, R.B.; Sery, T.; Spaeth, G.L. Pharmacokinetics of a 5-fluorouracil liposomal delivery system. Br. J. Ophthalmol. 1988, 72, 688–691. [Google Scholar] [CrossRef] [Green Version]
  195. Jin, S.; Ye, K. Nanoparticle-Mediated Drug Delivery and Gene Therapy. Biotechnol. Prog. 2007, 23, 32–41. [Google Scholar] [CrossRef]
  196. Occhiutto, M.L.; Maranhão, R.C.; Costa, V.P.; Konstas, A.G. Nanotechnology for Medical and Surgical Glaucoma Therapy—A Review. Adv. Ther. 2019, 37, 155–199. [Google Scholar] [CrossRef] [Green Version]
  197. Shao, T.; Li, X.; Ge, J. Target drug delivery system as a new scarring modulation after glaucoma filtration surgery. Diagn. Pathol. 2011, 6, 64. [Google Scholar] [CrossRef] [Green Version]
  198. Josyula, A.; Parikh, K.S.; Pitha, I.; Ensign, L.M. Engineering biomaterials to prevent post-operative infection and fibrosis. Drug Deliv. Transl. Res. 2021, 11, 1675–1688. [Google Scholar] [CrossRef]
  199. Shaunak, S.; Thomas, S.; Gianasi, E.; Godwin, A.; Jones, E.; Teo, I.; Mireskandari, K.; Luthert, P.; Duncan, R.; Patterson, S.; et al. Polyvalent dendrimer glucosamine conjugates prevent scar tissue formation. Nat. Biotechnol. 2004, 22, 977–984. [Google Scholar] [CrossRef]
  200. Bicket, A.K.; Szeto, J.; Roeber, P.; Towler, J.; Troutman, M.; Craven, E.R.; Khatana, A.; Ahmed, I.; Quigley, H.; Ramulu, P.; et al. A novel bilayered expanded polytetrafluoroethylene glaucoma implant creates a permeable thin capsule independent of aqueous humor exposure. Bioeng. Transl. Med. 2020, 6, e10179. [Google Scholar] [CrossRef]
  201. Preclinical Study: New Gore Glaucoma Drainage Implant-American Academy of Ophthalmology. Available online: https://www.aao.org/eyenet/academy-live/detail/preclinical-study-gore-glaucoma-drainage-implant (accessed on 7 November 2022).
  202. Gambini, G.; Carlà, M.M.; Giannuzzi, F.; Caporossi, T.; De Vico, U.; Savastano, A.; Baldascino, A.; Rizzo, C.; Kilian, R.; Caporossi, A.; et al. PreserFlo® MicroShunt: An Overview of This Minimally Invasive Device for Open-Angle Glaucoma. Vision 2022, 6, 12. [Google Scholar] [CrossRef]
Figure 1. Stages of wound healing and key markers following glaucoma filtration surgery.
Figure 1. Stages of wound healing and key markers following glaucoma filtration surgery.
Biomedicines 11 00657 g001
Table 1. Clinical, in vivo, and in vitro therapeutic agents and molecular targets for improving the success of glaucoma filtration surgery.
Table 1. Clinical, in vivo, and in vitro therapeutic agents and molecular targets for improving the success of glaucoma filtration surgery.
Clinical Studies
Therapeutic AgentMolecular TargetRoute/Type of DeliveryResultsPotential
Risks
Ref. No
Fluorouracil (5-FU)Inhibits DNA synthesisPO SC Inj. Anti-proliferative
 
Reduced filtration surgical failure
Hypotony maculopathy
 
Bleb leak
[39,40,41]
Mitomycin-C (MMC)Inhibits cell cycle, DNA replication, cell mitosis, and protein synthesisSC Inj.
 
Sponge
 
ST Inj.
Reduced scar formation
 
Increased bleb survival Rate
 
Maintained reduced IOP
Bleb leakage
 
Hypotony
 
Choroidal effusion
 
Endophthalmitis
[42,43,44,45,46]
Beta irradiationIncreases p53RadiationMinimized scar tissue formation
 
Maintained reduced IOP
Cataract formation
 
Keratopathy
[47,48]
Fibrinolytic therapyTissue plasminogen activatorIntracameralDissolved fibrin in blood clotsHyphemia
 
Profound hypotony
 
Anterior chamber flattening
[49]
Perfluoropropane (C3F8)Functional spacerSC Inj.Maintained reduced IOP [50]
HealaflowCross-linked sodium hyaluronateSurgical ImplantMaintained reduced IOP [48,51,52,53,54,55]
OlogenBiodegradable 3D COL-GAG copolymer matrix implantSurgical ImplantMaintained reduced IOP [56]
In vivo Studies
Therapeutic agentMolecular TargetRoute/type of DeliveryOcular Fibrosis
Model
ResultsPotential
Risks
Ref. No
Fluorouracil (5-FU)Inhibits DNA synthesisTopical Eye DropsRabbitAnti-proliferative
 
Reduced filtration surgical failure
Toxic to actively replicating tissue[57,58]
SMADOverexpress SMAD7
 
Reduces SMAD2, 3, 4
MicroRNAs (miRNA)RabbitReduced stromal fibrosis
 
Modulated α-SMA expression
[26,59,60]
Anti-SPARCSPARCNanoparticle-based-siRNAMouse
 
Rabbit
Less collagenous ECM
 
Smaller fibril collagen diameter
 
Improved surgical survival
[61,62]
PirfenidoneInhibits TGF-β
 
Reduces TNFɑ
Topical Eye DropsRats
 
Rabbits
Inhibited extracellular matrix deposit
 
Improved bleb survival
[63,64]
Anti-PDGF Subunit B agentsBinds PDGFARC126/127
 
E10030
MouseAnti-angiogenic
 
Inhibited tumor growth
[65,66]
Anti-VEGF AgentsInhibits VEGFSC Inj. (Bevacizumab)Rabbit
 
Mouse
Reduced IOP
 
Reduced angiogenesis
 
Reduced collagen deposition
Scleral blebbing[67]
MMC + SaratinInhibits cell cycle + prevents platelet aggregationSC Inj.
+
Topical application
RabbitIncreased bleb survival with a vascular bleb and tissue thinning [68]
Lysyl Oxidase-Like 2Collagen and elastin cross-linkingInhibitory monoclonal antibodyRabbitReduced pathologic angiogenesis, inflammation, and fibrosis [69]
MMPDegrades extracellular matrixTopical Inhibitor
 
SC IIlomastat
RabbitImproved bleb survival
 
Reduced IOP
 
Reduced scar tissue
[70,71]
MRTFFibroblast activationNanoparticle siRNARabbitDoubled bleb survival
 
Decreased conjunctival scarring
[72]
SAHAHistone modifierSC Inj.RabbitImproved bleb survival [73]
p53 inhibitorsTumor suppressor geneAAVRabbitReduced IOP
 
Prevented fibrosis proliferation
[74]
CDKN1BCell division pathway inhibitorTopical InhibitorRabbitReduced scar severity
 
Improved surgical outcomes
[75]
CyclosporineImmunosuppressiveTopical ApplicationRabbitEnhanced effectiveness of glaucoma drainage [76]
amniotic membraneAnti-inflammatory,
antifibrotic,
antiangiogenic
TransplantationRabbitImproved bleb survivalDelayed wound healing[77]
Perfluoropropane (C3F8)Functional spacerSC Inj.RabbitMaintained reduced IOP [78]
In vitro Studies
Therapeutic agentMolecular TargetCell TypeResultsRef. No
5-FUAntagonizes pyrimidine metabolismEpithelial
 
Fibroblast
Anti-proliferative
 
Inhibited fibroblast-mediated collagen contraction
[79]
MMCDNA crosslinking alkylating agentFibroblastInhibited fibroblast proliferation[80,81,82]
Beta IrradiationIncreased p53FibroblastDiminished fibroblast proliferation
 
Targeted ECM production
[83,84]
Anti-TGF-βBlocks TGFβFibroblastMitigated fibroblast transdifferentiation[85,86]
YAP/TAZActivates SMAD2/3
 
TGF-β1 and 2 mediators
FibroblastReduced scaring markers[87,88]
SPARCUpregulates TGFβ2FibroblastModulated ECM production and organization[89,90]
TGIF 1 and 2Repress SMAD-dependent signalingFibroblastAttenuated myofibroblast formation[91,92,93,94]
PirfenidoneInhibits TGF-β
 
Reduces TNFɑ
Epidural cell
 
Fibroblast
Inhibited extracellular matrix deposit
 
Decreased fibroblast proliferation, migration, and contraction
[95,96]
NintedanibInhibits PDGF, VEGF, and FGF receptorFibroblastPotent antifibrotic effect[97,98]
SaratinCollagen-Platelet interaction inhibitorPlateletLimited PDGF, TGFb, IGF, and EGF expression
 
Prevented platelet aggregation
[99]
MMPDegrades extracellular matrixFibroblastReduced collagen contraction, cell migration, and collagen production[100]
MRTFFibroblast activationFibroblastBlocked collagen matrix contraction[101]
p53 inhibitorsTumor suppressor geneFibroblastInhibited proliferation and DNA synthesis[102,103]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shao, C.G.; Sinha, N.R.; Mohan, R.R.; Webel, A.D. Novel Therapies for the Prevention of Fibrosis in Glaucoma Filtration Surgery. Biomedicines 2023, 11, 657. https://doi.org/10.3390/biomedicines11030657

AMA Style

Shao CG, Sinha NR, Mohan RR, Webel AD. Novel Therapies for the Prevention of Fibrosis in Glaucoma Filtration Surgery. Biomedicines. 2023; 11(3):657. https://doi.org/10.3390/biomedicines11030657

Chicago/Turabian Style

Shao, Christine G., Nishant R. Sinha, Rajiv R. Mohan, and Aaron D. Webel. 2023. "Novel Therapies for the Prevention of Fibrosis in Glaucoma Filtration Surgery" Biomedicines 11, no. 3: 657. https://doi.org/10.3390/biomedicines11030657

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop