Next Article in Journal
Supercritical CO2 Extraction and Tandem Mass Spectrometry of the Medicinal Plant Sagan Dalya (Rhododendron adamsii)
Previous Article in Journal
A Unified Approach to Synthesizing Four Linezolid Metabolites That May Cause Thrombocytopenia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Challenge of Corneal Ulcer Healing: A Novel Conceptual Framework, the “Triad” of Corneal Ulcer Healing/Corneal Neovascularization/Intraocular Pressure, and Avascular Tendon Healing, for Evaluation of Corneal Ulcer Therapy, Therapy of Neovascularization, Glaucoma Therapy, and Pentadecapeptide BPC 157 Efficacy

by
Sanja Masnec
1,2,
Antonio Kokot
3,*,
Tamara Kralj
1,
Mirna Zlatar
1,
Kristina Loncaric
1,
Marko Sablic
3,
Miro Kalauz
2,
Iva Beslic
2,
Katarina Oroz
1,
Bozana Mrvelj
1,
Lidija Beketic Oreskovic
1,
Ivana Oreskovic
1,
Sanja Strbe
1,
Borna Staresinic
1,
Goran Slivsek
1,
Alenka Boban Blagaic
1,
Sven Seiwerth
4,
Anita Skrtic
4 and
Predrag Sikiric
4,*
1
Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
2
Department of Ophthalmology, University Hospital Center Zagreb, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
3
Department of Anatomy and Neuroscience, School of Medicine Osijek, University of Osijek, 31000 Osijek, Croatia
4
Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(12), 1822; https://doi.org/10.3390/ph18121822
Submission received: 13 November 2025 / Revised: 24 November 2025 / Accepted: 26 November 2025 / Published: 28 November 2025

Abstract

To better address the challenge of corneal ulcer healing, with already available standard agents, and those recently introduced, such as stable gastric pentadecapeptide BPC 157, we introduced a novel conceptual framework—the “triad” of corneal ulcer healing↔corneal neovascularization↔intraocular pressure—and extended it to avascular tissues such as tendon. Within this framework, cytoprotection serves as the unifying principle, underscoring that therapeutic effects are not isolated but interconnected. Preclinical studies with BPC 157 therapy, as a cytoprotection agent, illustrate this integration. BPC 157 rapidly normalizes elevated intraocular pressure in glaucomatous rats, preserves retinal integrity, restores pupil function, maintains corneal transparency during ulcer or abrasion healing, and counteracts both corneal neovascularization and dry eye. In parallel, its consistent efficacy in tendon injury models highlights a cytoprotective specificity across avascular tissues. The cornea’s “angiogenic privilege,” preserved during healing and tendon recovery together, provides strong proof of concept. Furthermore, mapping standard therapeutic agents used for corneal ulcers, neovascularization, or glaucoma onto this triad, and linking them with tendon healing, reveals both shared pathways and inconsistencies across existing drug classes. Analyzed were the ascorbate, fibronectin, hyaluronic acid, metalloproteinase inhibitors, EGF, FGF, NGF, insulin, and IGF-1 (corneal ulcer healing), the antiangiogenic agents (endostatin, PAI-1, PEDF, angiostatin, TSP-1, TSP-2, IFN-α), corticosteroids, NSAIDs, cyclosporine A, anti-VEGF drops (treatment of corneal neovascularization), and alpha 2-agonists, beta-blockers, carboanhydrase inhibitors, muscarinic agonists, Rho-kinase inhibitors, and prostaglandin analogs (glaucoma). Taken together, these findings advance cytoprotection as a unifying therapeutic paradigm, with BPC 157 emerging as its first exemplar, and encourage further translational research toward clinical application.

1. Introduction

Better resolution of corneal ulcer healing with pharmacotherapy is a pertinent challenge. To this point, this review attempts to promote a new therapy way, based on the available standard agents [1,2], and those recently introduced (i.e., stable gastric pentadecapeptide BPC 157 [3]). This would be sharing particular interconnections, an essential “triad”, corneal ulcer/wound healing, corneal neovascularization, and intraocular pressure.
Recently, as a cytoprotective agent therapy [4,5,6,7,8], the stable gastric pentadecapeptide BPC 157 was reviewed in glaucoma [9], and in other tissues [10,11]. This was providing pleiotropic beneficial effects [12], a long-known conceptual characteristic of cytoprotective agents as pointed out in funding cytoprotection papers (i.e., [13,14,15,16,17,18,19,20,21]), and, especially, in the particulars of wound healing [22,23,24,25]. With this new conceptual perspective, for corneal ulcer healing, this therapy perspective was now reviewed compared to standard existing eye therapies (i.e., anti-VEGF agents, corticosteroids, EGF, NGF, and others) used in corneal ulcer healing, therapy of neovascularization, and glaucoma therapy. This was carried out using the mentioned novel “triad” approach (corneal ulcer healing↔corneal neovascularization↔intraocular pressure). Notably, this may be important, providing that the standard agents act in “fragments” and conceptually emphasize why these fragmented approaches fail to prevent cross-effects (e.g., anti-VEGF may impair ulcer healing [26], corticosteroids may worsen intraocular pressure [27,28], etc.).
To highlight the eye therapy in general, and corneal ulcer therapy, in particular, it may be a unified framework (“cytoprotection”) that conceptually links corneal ulcer healing, corneal neovascularization, and intraocular pressure regulation. This is a fresh perspective compared to the traditional fragmented approaches, where these phenomena are usually studied and treated in isolation [1,2]. Positioning them as a “triad” is a compelling way to highlight their interdependence. Likewise, this “triad” could hallmark the full efficacy and potential of the agent used in either eye therapy. Highly regarded in the literature, but never practically implemented, there is the concept of cytoprotection, as one of the most precise holistic concepts (i.e., innate cell protection against diverse noxious events toward original status, and therefore, integrated beneficial agents’ effects [13,14,15,16,17,18,19,20,21]).
While focusing on corneal ulcer healing, this review, based on the different agents’ effects and use in eye therapy (corneal ulcer, or corneal neovascularization, or increased intraocular pressure), considers the general evidence that corneal ulcer healing, corneal neovascularization, and intraocular pressure are interrelated [1,2,29,30,31,32,33,34], a “triad”, and together they strongly influence ocular integrity and visual outcome. As commonly acknowledged, they act through linked biological and mechanical pathways (inflammation→angiogenesis→altered aqueous dynamics/structural damage), so problems in one domain commonly affect the others. Thereby, not only in theory, but more in practice, for long-term ocular integrity (i.e., clear, structurally stable cornea) and optimal visual outcome (i.e., preservation of transparency and optic nerve function), conceptually, the corneal ulcer healing (e.g., ↑)↔corneal neovascularization (e.g., ↓)↔intraocular pressure (e.g., ↓) relation is integrated in one agent’s effect. At best, such balanced therapy simultaneously promotes epithelial/stromal repair, suppresses pathological neovascularization, and maintains or recovers physiological intraocular pressure. In a broader context, the particular aspect of that system (corneal transparency/avascularity), the potential of drugs and drug classes supposed to activate or inhibit the system by interacting depending on circumstances, with specific molecular targets, highlights the corresponding effect on other tissue healing, particularly those avascular (i.e., tendon) [4] as an “external validation axis”. Thereby, evidenced tendon healing should be regarded as a hallmark of the well-orchestrated effect [4] (see Figure 1).
Notably, this tendon/cornea avascular analogy as a clue could serve as an independent validation axis for agents that modulate angiogenesis and matrix remodeling [4]. Certainly, it departs from current therapy-specific terms of either ulcer healing, or the therapy of neovascularization, or the decrease in intraocular pressure [1,2,29,30,31,32,33,34], since the current eye therapy does not consider musculoskeletal disorders [1,2,29,30,31,32,33,34]. As a crucial advantage [4], defining the agent’s activity also by tendon healing (avascular analogy) may reveal a particular aspect of corneal ulcer healing itself. Furthermore, such additional (in)direct characterization of corneal ulcer healing [4] would exemplify positive and negative innate interconnections between corneal ulcer/neovascularization/intraocular pressure, revealing particularities of the “triad” as a print of the agent’s activity.
In this context, recently, we have attempted to implement the cytoprotection concept in eye therapy [9]. The evidence, still preclinical, was reviewed as a novel insight that the stable gastric pentadecapeptide BPC 157 therapy can recover glaucomatous rats [9], rapidly lower increased intraocular pressure to normalize intraocular pressure, maintain retinal integrity [35], and recover pupil function [36]. Accordingly, it recovers retinal ischemia [37]. Likewise, it heals corneal injuries (i.e., maintains transparency after complete corneal abrasion [38] and corneal ulceration [3], and counteracts dry eye after lacrimal gland removal [39] or corneal insensitivity [40]) (Table 1).
Practically, for eye therapy, particularly in the context of corneal ulcer healing, this conceptual cytoprotective approach can be beneficial. Notably, this may be for stomach ulcer~corneal ulcer [3]. Since 1979, the concept of cytoprotection has evolved from a breakthrough management strategy for stomach ulcers to a broader therapeutic approach, encompassing organoprotection (i.e., cytoprotection→organoprotection). It holds as a novel therapy, the maintaining and reestablishing original gastric integrity via the orchestrated protection of epithelial and endothelial cells. In practice, via cytoprotection agent application, the cytoprotection concept is proven and translated to other organ therapies (cytoprotection→organoprotection) [13,14,15,16,17,18,19,20,21]. An indicative analogy is prostaglandin analogs, i.e., latanoprost, key glaucoma therapy [41], as prostaglandins were the first mediators of cytoprotection as a concept of general (healing) significance, directly preventing epithelial necrosis that may arise in the stomach from the direct injurious effect of various agents’ applications, and thereby in other tissues as well [13,14,15,16,17,18,19,20,21]. Therefore, conceptually, there is a pleiotropic beneficial effect for cytoprotection agents [13,14,15,16,17,18,19,20,21], and for BPC 157, easily applicable, as a supposed cytoprotection mediator, as native and stable in human gastric juice, in particular [4,5,6,7,8,9,10,11,12,23,24,25,26]. Notably, recently, BPC 157 pleiotropic effects were reviewed as a therapy and safety key [4]. A special beneficial effect occurs by controlling and modulating angiogenesis and NO-system, targeting angiogenesis and NO’s cytotoxic and damaging actions, but maintaining, promoting, or recovering their essential protective functions, thus, cornea healing “angiogenic privilege” along with advanced tendon healing as a proof of the concept [4].
This cytoprotection translational evidence [4], taken together, should be crucial, especially in the context of corneal ulcer healing (corneal avascularity vs. corneal neovascularization; corneal healing vs. healing in other organs). From theory to practice, these cytoprotection terms should include all points mentioned, as noted with BPC 157 eye therapy. These were glaucomatous rats’ recovery [9,35], increased intraocular pressure promptly lowered and normalized [35], and the recovery of retinal integrity [35,37], pupil function [35,36], and retinal ischemia [37]. BPC 157 therapy maintained transparency while healing of corneal injuries [3,38,39,40], and counteraction of dry eye and corneal insensitivity occurred while maintaining tear production [39,40]. Linking with tendon healing assures the needed specificity of the therapy [4] (note, BPC 157 therapy’s beneficial effects are confirmed in several tendon injury models [42,43,44,45,46,47,48,49]).
These should be viewed in the context of a persistent attempt to develop more effective corneal management, particularly for corneal ulcers, given the numerous reviews that reanalyze all the particularities of corneal wound healing as a complex process, although, without combining with cytoprotection theory. Notably, this was without referring to the integration of corneal ulcer healing/corneal neovascularization/intraocular pressure for therapy’s purpose, or tendon healing as a hallmark of an orchestrated healing capacity. The general focus was on the significance of the corneal anatomy, the mechanisms of corneal epithelial and stromal wound healing, and endothelial dysfunction (i.e., one of the most common causes of corneal blindness) [50]. Then, the focus was on decreased corneal sensation that may indicate an underlying neurotrophic ulcer or herpetic ulcer [51,52], coordinated interaction of corneal cellular elements, a variety of cytoactive factors, components of the extracellular matrix (ECM), and biomechanical forces such as eyelid movement, and soluble factors known to play major roles in modulating these processes [1,33,53,54,55,56]. Likewise, with no integrating attempts, the other focus was on opposing neovascularization and the therapy [1,56,57,58,59]. In the same way, the additional sole focus was on the elevated intraocular pressure, which may be commonly present [29,30,60,61].
Therefore, corneal ulcer healing/corneal neovascularization/intraocular pressure, the “triad”, remains to be further redefined given the full effectiveness implications for the agents implicated in corneal healing, and corneal ulcer, in particular.
Moreover, as we recently elaborated [4], the credibility of these cytoprotection requirements is challenged by a common requirement for adverse effects. Likewise, there is widespread understanding that any of the therapeutic interventions have side effects. Contrarily, the concept holds that there is such general therapy involving all organs, together and in particular, that should not produce any harm on the other side. Cytoprotection is conceived as a mechanism that, in principle, should avoid collateral injury because it restores homeostasis rather than overriding physiology [13,14]. Conceptually, given that a cytoprotection agent, by definition, should not produce any adverse effect on account of its beneficial effects (although this is not the case with prostaglandins), the corneal ulcer healing/corneal neovascularization/intraocular pressure controlling relations should also link the avascular tendon. Thereby, it would depict the healing depending on timing and tissue specificity [4]. Thus, augmenting ocular cytoprotection with tendon-healing readouts provides an avascular tissue benchmark that clarifies an agent’s capacity to restore homeostasis in corneal ulcer disease, enabling more mechanism-driven translation [4].
Likewise, this implicates high effectiveness (i.e., BPC 157 therapy is effective in the 10 µg–10 ng/kg range). In toxicology studies, BPC 157 exhibited a negative limit test, 2 g/kg i.v. or i.g., without adverse effects in mice, and a lethal dose (LD1) was not achieved [4,5,6,7,8,9,10,11,12,23,24,25,26]. Later, it was effectively used in ulcerative colitis trials (phase II) without adverse effects [62,63], and quite recently, in knee pain [64] and interstitial cystitis [65]. Thus, together, these clinical human data can join the large range of beneficial effects of the BPC 157 therapy indicated by animal experiments, although the human studies performed so far are scarce, with limitations in some of them (i.e., they lacked a large sample size, ethnic variation, and a sham control group). However, these clinical studies effectively encompassed a wide range of investigations (for additional points in musculoskeletal disorders, see [66,67]). Consistently, there are similar beneficial results of BPC 157 therapy obtained after different application routes. Thus, such high applicability of the agent [4,5,6,7,8,9,10,11,12,23,24,25,26] (i.e., native and stable in human gastric juice for more than 24 h [68]), overwhelms the standard cytoprotective agents’ limited (prophylactic application only) applicability and activity [4,5,6,7,8,9,10,11,12,23,24,25,26]. This clearly shows the avoidance of pitfalls already indicated during the concept’s establishment [13,14,15,16,17,18,19,20,21], and it attempts [4,5,6,7,8,9,10,11,12,23,24,25,26] to realize the concept’s tools.
All of these points may be supported by its special interaction with various molecular pathways [69,70,71,72,73,74,75,76,77,78,79,80]. This includes, as a major point, the NO-system [70,71,72] as a whole, counteraction of the adverse effect of either NO-blockade or NO-overactivity, demonstrated in more than 80 targets (i.e., hypertension, hypotension, thrombosis, bleeding) investigated [81,82,83,84,85].
At least partly, this explains the cytoprotective ability of the pentadecapeptide BPC 157 to further maintain and upgrade endothelium integrity and functioning (for review see [4]), particularly focused on minor vessels during noxious procedures, rapid upgrading, and collateral pathways’ activation to substitute the function of the failed major blood vessels [4]. Illustratively, this likely occurred in glaucomatous rats, where one episcleral vein remained and took over the failed function of all the episcleral veins, amid rapid lowering of increased intraocular pressure as proof [9,35].

2. Corneal Ulcer Healing Therapy in Terms of “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Beyond the well-documented evidence of diverse etiologies of corneal ulceration, current therapy is largely etiology-specific. For infectious ulcers, antibiotics are used against bacteria, antivirals against viruses (predominantly herpes simplex), antifungal drugs for fungal ulcers, and combined antiseptic/anti-amoebozoal therapy for parasitic infections such as Acanthamoeba. For non-infectious causes, treatment is directed at the underlying condition, such as lubricating drops for dry eye or immunosuppressants for autoimmune disorders [86]. In parallel, numerous trials have aimed to enhance corneal wound healing through topically applied agents, most commonly ascorbate, fibronectin, hyaluronic acid, metalloproteinase (MMP) inhibitors, EGF, fibroblast growth factor (FGF), NGF, insulin, and insulin-like growth factor-1 (IGF-1). However, as already emphasized, despite these efforts, no analysis has yet addressed the integrated relationship between corneal ulcer healing, corneal neovascularization, and intraocular pressure, nor has this been linked to healing in other avascular tissues (e.g., tendon), which could serve as a valuable comparative model and a healing highlight.

2.1. Ascorbate

A considerable number of manuscripts showed corneal benefit from topical/systemic ascorbate and corneal ulcer healing. Ascorbate speeds epithelial re-closure, and lowers inflammatory cell influx and neovascularization in many models. Thus, (healing + reduced neovascularization) is supported in multiple in vivo corneal models (alkali and thermal burns, epithelial debridement, suture/cauterization models); topical ascorbate (commonly 10% or other concentrations depending on the model) accelerates epithelial repair or reduces stromal ulceration and—importantly—reduces corneal neovascularization in experimental models [87,88,89,90,91,92,93,94,95,96].
Furthermore, considering (healing + reduced neovascularization + reduced intraocular pressure), the published experimental and clinical literature suggests either no effect or, more often, an intraocular pressure-lowering/intraocular pressure-modulating effect of ascorbate (including IV or very high oral doses producing transient osmotic intraocular pressure drops) [84,88,89,90,91,92,93,94,95,96].
Finally, considering the final extension (healing + reduced neovascularization + reduced intraocular pressure + tendon healing), ascorbic acid/vitamin C accelerates tendon healing associated with early angiogenesis or changes in angiogenic markers [97,98,99,100,101,102,103]. The preclinical evidence is consistent in rat, rabbit, and chicken models + in-vitro tenocyte/tendon-derived stem cells work, as reported acceleration of tendon repair besides earlier angiogenesis includes increased fibroblast/tenocyte activity, greater type I collagen production, larger collagen fibril diameter, better collagen organization, and improved biomechanical strength.

2.2. Fibronectin

Fibronectin is a large extracellular matrix glycoprotein with a molecular weight of approximately 500–600 kDa. It interacts with integrins—transmembrane receptor proteins—as well as other matrix components, including collagen, fibrin, and heparan sulfate proteoglycans such as syndecans. Fibronectin healing of corneal ulcer in rabbits and patients (purified from autologous plasma by affinity chromatography and administered topically) [104,105,106,107,108,109,110,111,112,113,114,115,116,117] was ascribed to facilitation of corneal epithelial migration. Aiding surface re-epithelialization and decreasing corneal ulceration was ascribed to a prominent fibronectin–fibrinogen matrix, which remained on the surface of fibronectin-treated corneas. There is no direct evidence demonstrating that topically applied full-length plasma fibronectin induces de novo vessel formation in an otherwise normal cornea. However, considering corneal neovascularization, fibronectin is recognized as a pro-angiogenic molecule. Multiple lines of evidence support a causal role of fibronectin in ocular angiogenesis, implicating fibronectin itself, its domains or fragments, and its receptors in the regulation of angiogenic processes within the cornea and other ocular tissues [118,119,120].
There, fibronectin-1 is consistently elevated in vascularized corneas, implicating a pathologic neovascularization process, particularly via endothelial and immune cell interactions [121]. Moreover, considering fibronectin (corneal healing (↑) + corneal neovascularization (↑)) to intraocular pressure, many studies (in)directly link fibronectin/fibronectin fibrils to increased intraocular pressure or trabecular meshwork dysfunction. The increased density of fibronectin fibrils is commonly thought to increase intraocular pressure by altering the compliance of the trabecular meshwork [122,123,124,125,126,127,128]. In rats, fibronectin alone can chronically elevate intraocular pressure and induce neurodegenerative changes given a single intracameral injection of biodegradable poly(lactic-co-glycolic acid (PLGA) microspheres loaded with fibronectin [129].
Finally, fibronectin/tendon healing/angiogenesis is consistent with a lack of direct evidence for fibronectin (corneal healing (↑) + corneal neovascularization (↑) + intraocular pressure (↑)), since there is no evidence that direct application of fibronectin alone (i.e., exogenous fibronectin as the sole therapeutic agent, without concurrent growth factors, cells, scaffolds or gene delivery) is sufficient to produce functional tendon healing and angiogenesis in vivo.
However, toward improved tendon healing is the evidence that bFGF/PDGF and other biologics increase fibronectin deposition in the provisional matrix and promote revascularization/angiogenesis during tendon healing [130].

2.3. Sodium Hyaluronan, Hyaluronic Acid

Hyaluronic acid is a naturally occurring glycosaminoglycan that, by virtue of its viscosity, elasticity, and other rheological properties (particularly for its biocompatibility), acts as a lubricating and shock-absorbing fluid in skin and joints and as an ocular lubricant [131]. The supportive background goes with hyaluronic acid as a major component of the extracellular matrix, which is supposed to play a key role in tissue regeneration, inflammation response, and angiogenesis, which are phases of wound repair [132]. Consistently, hyaluronic acid successfully accelerates re-epithelialization of corneal wounds in both animal and human studies [133,134,135,136,137,138,139,140,141,142].
As an additional important point, hyaluronic acid did not induce corneal neovascularization in rabbits [143], and in rat models of alkali-induced corneal burns, cross-linked and standard linear hyaluronic acid hydrogels, delivered via in situ forming hydrogels, resulted in faster re-epithelialization, lower stromal inflammation, and significantly reduced neovascularization [144].
On the other hand, the viscoelastic properties of hyaluronic acid on liquid connective tissue have been proposed for the treatment of tendinopathies, and tendon healing combined with the improved angiogenesis [145,146,147,148,149].
Contrarily, thought to be a large molecule, hyaluronic acid may impair trabecular meshwork outflow by clogging channels, thereby reducing aqueous humor drainage. Hyaluronic acid increased intraocular pressure in both animal and clinical settings [150,151,152]. Illustratively, acute intracameral injection of 1% hyaluronic acid induced a twofold increase in intraocular pressure in rats [150].

2.4. MMP Inhibitors

The significance of MMP inhibitors is tied to the general importance of the MMP family, which consists of enzymes involved in the degradation of the extracellular matrix in tissues. In the skin, keratinocytes and fibroblasts express various MMPs during healing [152]. Similarly, corneal epithelial cells and keratocytes secrete MMPs during healing following various corneal injuries. Overexpression in infectious keratitis (bacterial, fungal, viral, Acanthamoeba) occurs with driving collagen degradation, stromal melting, thinning, and potential perforation [153]. Moreover, there is interleukin (IL)-1β—amplified MMP production by corneal fibroblasts and tissue destruction even after pathogens are eradicated [154].
Thus, as an MMP inhibitor, doxocycline is illustrative. At sub-antimicrobial doses, doxycycline inhibits collagenases and MMPs in vitro. Systemic administration in rabbit models significantly reduced corneal ulcer incidence—from 85% to 9% after alkali burn ulceration—and halved perforation rates in Pseudomonas ulcers [155]. Likewise, a similar beneficial effect on corneal ulcer healing occurred with other MMP inhibitors [156,157,158]. Additional experimental agents include ascorbic acid, epigallocatechin gallate (EGCG) (green tea polyphenol), urokinase plasminogen activator (uPA)-plasmin inhibitors, corticosteroids, and even tumor necrosis factor (TNF)-α blockers like infliximab, all investigated for indirect or direct MMP-suppressive or anti-collagenolytic effects [154]. A particular point occurs with galardin, as a hydroxamate broad-spectrum MMP inhibitor with potential in preclinical settings—since concerns considering MMP-system general activity include impaired normal wound healing, tumor-related effects, and other off-target impacts [153].
MMP activity promotes corneal angiogenesis. Thereby, MMP inhibitors, in principle, inhibit corneal angiogenesis [158]. However, it may be an opposite potential theoretical point for metalloproteinase inhibitors. MMP-12, a macrophage-associated protease, inhibits corneal inflammation and neovascularization by regulating CCL2/CCR2 chemokine pathways, based on murine injury models [159,160]. Thus, inhibition of MMP-12 might theoretically worsen inflammation and neovascularization—though such induced corneal neovascularization via inhibition has not been directly tested [161].
On the other hand, systemic doxycycline (a broad-spectrum gelatinase inhibitor) weakened rat Achilles tendons post-injury, indicating that inhibiting necessary MMP activity early may impair biomechanical healing [162].
Commonly, MMPs support intraocular pressure homeostasis in the trabecular meshwork. In the trabecular meshwork (TM) of the eye, endogenous MMPs (e.g., MMP-1, MMP-2, MMP-3, MMP-14) regulate ECM turnover, thereby maintaining aqueous humor outflow and stabilizing intraocular pressure [163]. An imbalance favoring MMP inhibition (e.g., excessive TIMP activity) contributes to ECM accumulation, increased outflow resistance, and elevated intraocular pressure in models of glaucoma and ocular hypertension [163,164,165].

2.5. EGF

Since the very beginning, EGF has shared general significance with other growth factors, such as bFGF, PDGF, and VEGF, which are endogenously derived and are thought to play a role in the natural history of healing, having considerable therapeutic potency. Illustrating their presentation (i.e., in tear fluid [166], corneal epithelium [167]) and consequent potential activity as a general point, an instructive emphasis was made in gastrointestinal research; these relatively large peptides are active in ng quantities, and their molar potency is 2–7 million times superior to cimetidine-like drugs [168,169,170]. This might be related to the fact that these growth factors, i.e., in duodenal ulcer healing, stimulate with varying potency virtually all the cellular elements needed for ulcer healing, e.g., epithelial cell proliferation and migration by EGF > bFGF > PDGF, fibroblast proliferation by bFGF > PDGF, and angiogenesis by VEGF > bFGF >> PDGF >> EGF [168,169,170]. Thereby, in particular for corneal ulcer healing, in analogy with skin healing [171], activation of EGFR in corneal cells has been shown to be crucial for the regeneration of the epithelium, stroma, and endothelium after corneal injury. However, the molecular mechanisms involved remain largely unclear beyond what is known in skin tissue [167]. EGF promotes the synthesis of specific proteins and supports the proliferation and differentiation of corneal epithelial cells, keratocytes, and endothelial cells. Moreover, the EGF family of growth factors plays a key role in stimulating lacrimal gland secretion. EGF and EGF-like proteins also interact with neurotransmitter-activated pathways, enhancing both the volume and composition of tear production [172,173]. Thus, since the very beginning, there has been a considerable number of reports on EGF on corneal ulcer healing in animals [171,174,175,176,177,178,179,180] and humans [181,182,183,184,185].
Contrarily, EGF enhances and induces corneal neovascularization [186,187,188].
There is no evidence that exogenous EGF application affected intraocular pressure or caused an increase in intraocular pressure [189]. EGFR expression increases in response to intraocular pressure elevation upregulation in astrocytes, linked with NOS-2 and MMP expression, potentially contributing to neurodegeneration [190].
Finally, considering the skin/cornea analogy, the evidence that exogenous administration of recombinant human epidermal growth factor (rhEGF; also called nepidermin) accelerates wound re-epithelialization, angiogenesis, fibroblast proliferation, and collagen deposition is confronted with the fact that free EGF is unstable. Thus, as rapidly degraded by proteases, and cleared, therefore, intelligent delivery carriers are essential for therapeutic efficacy [191,192,193,194]. However, exogenous EGF (human recombinant EGF or hrEGF) (intralesional application) was applied and reported improved tendon/tendon-repair histology with increased vascularity [195,196], providing histological and immunohistochemical (vascularity, cellularity, collagen organization), while biomechanical improvements were mixed/absent (histology improved but not always tensile strength).

2.6. FGF

For FGF, the corneal ulcer healing–corneal neovascularization–intraocular pressure relation, and finally, tendon healing as a hallmark, should be regarded with complex evidence. FGF has been given essential importance as a family of cell signaling proteins, as they are implicated in mediating various processes, such as angiogenesis, wound healing, metabolic regulation, and embryonic development, through their specific receptors. FGF can stimulate angiogenesis and proliferation of fibroblasts, and it is a powerful angiogenesis factor. Twenty-three subtypes have been identified and divided into seven subfamilies. Therefore, there is a possible application in wound healing with good therapeutic effects, i.e., topically to diabetic foot ulcer, with a particular focus on FGF-1, FGF-2, FGF-4, FGF-7, FGF-21, and FGF-23 [197].
Likewise, along with reduced inflammatory cytokines (IL-6, TNF-α, MMP-2/9), increased anti-inflammatory IL-10 and antioxidant SOD-1, and improved tear production and nerve regeneration, corneal ulcer healing is consistently established with topical FGF application in several models, in animal and human studies, including topical recombinant human FGF-21, FGF-7, FGF-2, and recombinant human bFGF [198,199,200,201]. Illustratively, treatment with bFGF alone accelerated epithelial healing, and the combination with CCP (an antioxidant enzyme) further enhanced closure rates and reduced oxidative damage [202]. A comparative evaluation in rabbits that assessed recombinant human EGF versus basic FGF on corneal epithelial wound healing found that bFGF promoted epithelial cell proliferation and migration, similar to EGF, with enhanced healing dynamics [203].
Contrarily, along with increased VEGF-A, C, and D expression, amplifying angiogenic signaling cascades, FGF-2 (bFGF) is one of the most potent angiogenic factors in the cornea [56]. Following corneal injury, it is significantly upregulated and directly stimulates corneal vascular endothelial cells, promoting both blood and lymphatic vessel formation [204]. Illustratively, in classic corneal micropocket assays using pellets, FGF-2 alone reliably induces both angiogenesis and lymphangiogenesis in a dose-dependent manner [205] as well as in corneal suture and HSV keratitis [206,207].
Finally, considering FGF’s influence on intraocular pressure, in young chicks, intraocular injections of IGF-1 plus FGF-2 induced dose-dependent increases in intraocular pressure, along with ocular enlargement, anterior chamber shallowing, and lens thickening [208].
On the other hand, stimulation of angiogenesis is commonly thought to be essential for FGF therapy in tendon injuries, where the increase in both cellularity and vascularization corresponded with earlier functional recovery [209,210,211,212,213,214,215]. However, the effectiveness and delivery strategies are based on the scaffold-based release systems or cell/gene vectors [216].

2.7. NGF

At the general level, there is considerable significance of NGF, as a soluble protein produced by and acting upon many different cells located in the nervous, endocrine, and immune systems. It stems from exerting a critical role on epithelial cells and fibroblasts under normal and pathological conditions. NGF is constitutively expressed in human and rat corneas; all major corneal cells—including epithelial, stromal, endothelial, and limbal stem cells [217,218,219]. Consistently, along with promoting epithelial cell proliferation/migration, enhancing nerve survival and regeneration, regulating inflammation, facilitating fibroblast activity and angiogenesis, and restoring tear function, topical NGF significantly accelerated epithelial and stromal healing in animal models and human organ culture, restoring corneal integrity through induction of receptor-mediated signaling [219]. As a particular point, NGF enhances the expression of MMP-9 and cell migration in the epithelium, which are also key to corneal neovascularization pathogenesis [217].
However, to date, induction of corneal neovascularization has not been reported in clinical and preclinical studies specific to NGF in the cornea [220,221,222,223]. Indicatively, in human patients treated with topical NGF (murine or rhNGF), healing occurred without evidence of neovascularization—even in inflammatory and melting ulcers otherwise prone to vascular ingrowth [224].
There is no study demonstrating that applying NGF lowers an already-elevated intraocular pressure. Studies applying NGF to the eye (i.e., topical recombinant human NGF (rhNGF) or through conjunctival or retrobulbar administration) consistently demonstrate neuroprotective effects on retinal ganglion cells (RGCs) and confirm good safety and tolerability in humans. These investigations typically employed elevated intraocular pressure models, showing that NGF preserves RGC integrity despite increased intraocular pressure, or they monitored intraocular pressure as a safety parameter and found no NGF-induced reduction in intraocular pressure [225,226,227,228,229,230,231]. Several preclinical glaucoma models (e.g., induced ocular hypertension in rats) showed that topical or intraocular NGF administration did not cause further intraocular pressure elevation; instead, it promoted retinal ganglion cell survival, inhibited apoptosis, and supported functional recovery [226]. An open-label clinical pilot treated three progressive glaucoma patients with murine NGF eye drops (200 μg/mL, four times daily for seven weeks) and observed improvements in visual acuity, contrast sensitivity, visual field, and electrophysiological measures—with no reported intraocular pressure worsening [227].
Beyond its well-known neurotrophic actions, NGF also plays roles in non-neural wound healing [232]. In skin and oral wound models, it promotes angiogenesis via VEGF-Akt-NO signaling pathways, and NGF participates in angiogenesis and myofibroblast differentiation during tissue repair [228].
Yet, there is no study that has shown the application of NGF alone (e.g., local NGF injection or topical NGF) to directly produce tendon healing in an experimental or clinical model. However, specifically, after tendon injury, NGF expression in tenocytes and surrounding nerve fibers increases, promoting reinnervation and sensory nerve ingrowth via TrkA signaling [233]. NGF (10 µg given via osmotic pump over 7 days) improves ligament healing [234] by promoting reinnervation and angiogenesis and producing scars with enhanced mechanical properties.

2.8. Insulin (and the IGF Axis)

Insulin promotes corneal epithelial cell migration, proliferation, and nerve recovery largely via PI3K–Akt signaling [235,236]. A considerable number of studies supported the corneal ulcer healing effect in in vitro and animal work, and human application [235,237,238,239,240,241,242,243]. Regardless of long-term safety and optimal formulations remaining understudied, reported topical use has been well tolerated in published series with minimal systemic effects [244]. This is evidence that topical insulin/IGF signaling can aid corneal nerve regeneration—relevant in neurotrophic keratopathy [245].
Considering the influence on corneal neovascularization, topical insulin promotes epithelial healing but has not been commonly reported to cause corneal neovascularization in human series. Multiple human case series and small trials report faster epithelial closure with topical insulin and few immediate vascular complications, although most studies are small and with short follow-up [242]. Moreover, topical application of insulin encapsulated by chitosan-modified PLGA nanoparticles alleviates alkali burn-induced corneal neovascularization [246].
On the other hand, as a particular point, insulin (native) and soft-tissue wound healing are consistently established, as multiple animal and human studies show topical/local insulin accelerates wound closure and upregulates angiogenic mediators (VEGF, eNOS, SDF-1α) in cutaneous wounds—mechanistically relevant to tendon repair because angiogenesis supports early healing [247]. Moreover, there is a direct effect of insulin on tendon cell effects. At least in vitro, insulin can affect tendon progenitor/tenocyte proliferation and differentiation [248]. Clinical case series and small randomized trials using topical insulin for corneal epithelial healing (typical regimens ~0.5 U/drop QID or diluted solutions) report no change in IOP during follow-up and no systemic glycemic effects in most reports.
Animal wound-healing studies with topical insulin similarly report no intraocular pressure change [249,250]. Systematic reviews and meta-analyses of cohort studies report an increased risk of open-angle glaucoma in people with diabetes (pooled RRs ≈ 1.3–1.5). This is one of the strongest and most reproducible epidemiologic signals [251].
On the other hand, the evidence of insulin and the IGF axis promoting corneal epithelial cell migration, proliferation, and nerve recovery largely via PI3K–Akt signaling [234] should be seen with evidence that IGF-1 alone could not increase corneal ulcer healing and had only an effect combined with substance P (which is also ineffective when given alone) [252,253]. Furthermore, in a rat model of corneal neovascularization, suppression of IRS-1—a downstream mediator of IGF-1 signaling—led to inhibited vessel growth, implying that IGF-1/IRS-1 signaling contributes to corneal angiogenesis [254]. In the cornea, IGF-1 is listed among several angiogenic mediators capable of disrupting corneal avascularity when dysregulated [57].
Considering the influence on intraocular pressure, IGF-1 can be responsible for the increased intraocular pressure (at least in a synergistic mechanism). Namely, daily intraocular pressure of IGF-1 and FGF-2 caused dose-dependent increases in intraocular pressure (chick models), although given alone, they are both ineffective [255].
On the other hand, the beneficial effect of IGF-1 on tendon healing is focused on several reviews (i.e., [256]) postulated in rat and equine studies, using local application [257,258,259,260], and in humans [261]. However, some points remained, both in animal studies (i.e., no gross histologic difference reported; biomechanical failure load not clearly improved [257]) as well as in clinical trials (not improved tendon synthesis, structure, or patient-reported outcomes beyond exercise alone [262]).

2.9. Summary of Corneal Ulcer Healing Therapy in Terms of “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Summarizing, the majority of available evidence for several corneal wound healing agents remains preclinical. This is particularly true for ascorbate, fibronectin, hyaluronic acid, MMP inhibitors, FGF, and insulin/IGF-1, where supportive data largely derive from animal models, pilot studies, or small uncontrolled clinical series [242,263,264,265]. Only NGF (NGF, cenegermin) [266] and, to a lesser extent, EGF [267] and hyaluronic acid [133] have been tested in larger randomized clinical settings with regulatory approval achieved for NGF (Table 2). Note, Table 2 concisely summarizes the key topical agents trialed to enhance corneal wound healing [133,242,264,265,267,268,269,270], along with their mechanisms, representative PubMed citations (PMID/DOI), and evidence levels.
Importantly, no study to date has evaluated these therapies within an integrated experimental framework that considers the dynamic interplay of corneal ulcer healing, corneal neovascularization, and intraocular pressure [1,2], the corneal ulcer healing/corneal neovascularization/intraocular pressure “triad”, or links ocular outcomes to avascular-tissue (tendon) healing as a validation axis. On the other hand, most preclinical effects of these agents have firmly established the framework of corneal ulcer, including various mechanisms and multiple factors, and different standpoints to approach corneal ulcer healing. Therefore, these open the possibility for novel attempts even outside of the current frame [1,2]. Consequently, there is an additional, yet resolving, and novel approach, and the novel BPC 157 evidence, also preclinical. Thereby, it can be acknowledged that BPC 157 therapy accordingly arrives with healing corneal ulcer, counteraction of corneal neovascularization, and counteraction of the increased intraocular pressure [3,9,35,36,37,38,39,40], linking corneal phenomena to avascular tendon healing [42,43,44,45,46,47,48,49], as a first exemplar framing of corneal ulcer healing, corneal neovascularization, and intraocular pressure as a unified “triad” within a unified cytoprotection framework.
To implement the cytoprotection concept in eye therapy, in particular in corneal ulcer healing, perceiving BPC 157 corneal ulcer healing capability as a whole was carried out, respecting corneal healing agents as a class. Commonly, comparing corneal ulcer healing with the “triad” (corneal healing/corneal neovascularization/intraocular pressure relation) and healing in another avascular tissue like tendon, under a cytoprotection umbrella, these agents, ascorbate, fibronectin, hyaluronic acid, MMP inhibitors, EGF, FGF, NGF, and insulin/IGF-1, all revealed the particular “triad” relations, and a connection or no connection with tendon healing. These differences, now revealed, with the caveat that some direct evidence may be lacking, appeared to be likely specific to any of these agents’ activities organization (Table 3) (e.g., ↑ healing = beneficial, ↓ neovascularization = beneficial, ↑ intraocular pressure = harmful).
There, as an indicative point, using the application of the agents as a proof, there is ascorbate, along with the noted particular BPC 157 relation, where the corneal ulcer ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↑-relation occurred as a shared general most favorable effect. These appear to be close to the insulin course (corneal ulcer ↑ ↔ corneal neovascularization not affected (0) or ↓ ↔ intraocular pressure not affected (0), and tendon healing ↑) as well as to NGF (NGF corneal ulcer healing occurs as a sole corneal ulcer ↑, given non-affected (0), the corneal neovascularization and intraocular pressure, and likely ↑ tendon healing (given the noted ligament healing)). Some outcomes from such a course occurred with hyaluronic acid (corneal ulcer ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↑ and tendon healing ↑). Contrarily, different, perilous interrelations occurred, likely specific, for fibronectin (corneal ulcer ↑ ↔ corneal neovascularization ↑↔ intraocular pressure ↑), MMP-inhibitors (corneal ulcer ↑ ↔ corneal neovascularization ↓↔ intraocular pressure ↑ and tendon healing ↓), EGF (corneal ulcer ↑ ↔ corneal neovascularization ↑↔ intraocular pressure not affected (0), and ↑ tendon healing), and FGF (corneal ulcer ↑ ↔ corneal neovascularization ↑ ↔ intraocular pressure ↑, and ↑ tendon healing). IGF-1 is different (corneal ulcer healing not affected (0) (application alone)↔corneal neovascularization ↑ ↔ intraocular pressure ↑, and ↑ tendon healing).
Thus, from the viewpoint of the adequate and safe healing of the corneal ulcer, with “triad” presentation, and connection with tendon healing, as an additional key, the so far acknowledged large range of the framework of corneal ulcer, including various mechanisms and multiple factors, and different standpoints to approach corneal ulcer healing, should be further clarified and specifically pointed out. Whatever the specific mechanism behind this, some points should be favored (i.e., ascorbate) or presumed (hyaluronic acid, insulin, NGF), while others could be possibly discarded (i.e., fibronectin, MMP-inhibitors, EGF, FGF, and IGF-1), as a triad-based approach may better capture therapeutic efficacy and safety in the complex environment of ocular surface disease.
To support these favorable points, BPC 157 therapy for corneal ulcer healing is also combined with maintaining transparency and counteracting corneal lesions after complete corneal abrasion, dry eye after lacrimal gland removal, or cornea insensitivity after topical ophthalmic anesthetics, as well as counteracting the decrease in the tear volume [3,9,35,36,37,38,39,40]. In a broader wound context, i.e., skin wound healing, regularly used to support the beneficial effect of growth factors on corneal ulcer healing, BPC 157 therapy, given alone, without any carrier addition, unlike growth factors, as reviewed, heals both external and internal wounds [22,23,24,25]. These include also in diabetic animals [271,272] a beneficial effect, whatever the route of application. Moreover, as an important analogy for corneal lesions healing, BPC 157 wound healing is a particular one [22,23,24,25]. This can be suggested, providing simultaneous healing of different tissues, as noted with both internal and external fistula healing, and various anastomosis healing, given the specific beneficial outcome, toward the reestablishment of the original presentation (i.e., fistula closure, concomitant damages counteracted, healed intestinal anastomosis, and adhesions counteracted) [24,25]. Note, without therapy, some of the fistulas could be lethal in rats [24]. Likewise, the tendon healing improvement, in vivo and in vitro [42,43,44,45,46,47,48,49,76,77], is well-grounded and reviewed [4,10,11,66,67,273], given the beneficial effect after tendon transection, and detachment from bone or muscle [42,43,46,47].

3. Corneal Neovascularization

3.1. The Antiangiogenic Agents in Terms of the “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

The antiangiogenic agents include endostatin and endostatin analogs (neostatin, canstatin, tumstatin), plasminogen activator inhibitor-1 (PAI-1), and serine protease inhibitors, retinal pigment epithelium-derived factor (PEDF), angiostatin, thrombospondin 1,2 (TSP-1, TSP-2, respectively), and interferon (IFN)-α [1,56,57,58,59].

3.1.1. Endostatin

Suppressed corneal neovascularization accompanied by enhanced corneal healing outcomes was consistently reported in numerous studies which have evaluated the application of endostatin (i.e., in its native protein form, engineered or modified variants, or via gene delivery) in corneal ulcer and corneal injury models [274,275,276,277,278].
No single primary paper was identified that directly and explicitly reports the application of endostatin (in a corneal/ulcer model) inducing an increase in intraocular pressure while simultaneously counteracting corneal neovascularization.
Counteraction of corneal neovascularization appeared in ocular gene-therapy/intravitreal/subretinal studies that express endostatin and/or angiostatin (RetinoStat® and related preclinical work) [279,280,281,282]. The preclinical papers report effective suppression of choroidal neovascularization in laser corneal neovascularization models and no persistent, vector-attributable increases in intraocular pressure in the preclinical safety programs (intraocular pressure was explicitly monitored in the Binley safety/biodistribution studies) [280,281,282].
No paper directly tested the application of exogenous endostatin (local injection into tendon or systemic administration) and reported a measured decrease in tendon healing as the primary outcome. The closest direct experimental evidence [283,284,285] supports that excess endostatin can delay skin wound healing [283,284] and that endostatin is produced in tendon and regulated by mechanical load [285]. These findings provide plausible mechanistic support that exogenous endostatin could impair tendon healing via anti-angiogenic and matrix-modifying actions—but they are not direct tendon-intervention studies [283,284,285].

3.1.2. PAI-1

There is no study in which the recombinant PAI-1 (or other exogenous PAI-1 preparations) was applied to the cornea (topically, subconjunctivally, intracorneally, intrastromally, intravitreally, or systemically) and shown to reduce corneal neovascularization. On the other hand, the supporting evidence is obtained in context/dose dependence in ocular compartments other than the cornea. The expression of PAI-1 mRNA was analyzed in human and murine choroidal neovascularization by RT-PCR. The influences of increasing doses of recombinant PAI-1 were evaluated by daily intraperitoneal injections in PAI-1 (-/-) and wild-type animals with a model of laser-induced corneal neovascularization [286]. PAI-1 is pro-angiogenic at physiological concentrations and anti-angiogenic at higher concentrations [287]. PAI-1 facilitates retinal angiogenesis in a model of oxygen-induced retinopathy [288].
The plasminogen/PA system in corneal ulceration and wound healing is largely reviewed [1,289]. However, only one in vivo study, carried out with subconjunctival injection of recombinant Serpine1 (PAI-1) in streptozotocin-diabetic mice, shows that exogenous (PAI-1/Serpine1) applied to the eye accelerated corneal epithelial wound closure [290]. Further supporting studies are in vitro demonstrations that exogenous PAI-1 promotes adhesion and chemotactic migration of human corneal epithelial cells—mechanistic support for the in vivo findings [291]. Likewise, exogenous PAI-1 enhances keratinocyte migration and protects against plasminogen-induced detachment—supportive evidence from epidermal models (not cornea) [292].
There is no study to show that exogenous PAI-1 administration by itself (topical, intracameral, intravitreal, subconjunctival, or systemic) would increase intraocular pressure in animal models or humans. However, considering combining PAI-1 with increased intraocular pressure, PAI-1 contributes to glaucoma pathogenesis; the evidence includes the elevated PAI-1 in eyes with glaucoma/aqueous humor, and PAI-1 expression in anterior-segment tissues that can affect outflow; thus, the plasminogen/plasmin system (and tPA) modulates outflow/intraocular pressure [293,294,295,296].
Although there is no study in which the application of PAI-1 (plasminogen-activator-inhibitor-1) alone (topical, local or systemic) on tendon healing was tested, PAI-1 is associated with fibrotic adhesions after tendon injury, while genetic deletion or therapeutic inhibition of Serpine1 (PAI-1) reduces adhesions and improves remodeling [297,298,299].

3.1.3. PEDF

Exogenous PEDF (full-length protein or PEDF-derived peptides/fragments) promotes corneal epithelial/limbal wound healing and/or inhibits corneal neovascularization [300,301,302,303,304].
Direct experimental evidence that PEDF application increases outflow resistance was provided by Rogers et al., 2013 [305], who showed that purified PEDF decreases outflow facility in enucleated mouse eye perfusions and increases transendothelial electrical resistance of Schlemm’s canal endothelial cells. Thus, PEDF application could raise intraocular pressure (because decreased outflow facility→higher intraocular pressure). Moreover, PEDF is present and is differentially abundant in the aqueous humor in inflammatory and glaucomatous states [306,307,308].
Application of PEDF or PEDF-derived peptides promotes tendon healing/tendon regeneration (or closely related tissue-regenerative effects) [309,310,311]. Illustratively, topical/local delivery of a PEDF-derived short peptide in an alginate hydrogel promoted Achilles tendon regeneration in a rat full-thickness injury model (improved histology, better collagen alignment, and increased tensile strength) [309].

3.1.4. Angiostatin

There is compelling evidence for the application of angiostatin reducing corneal neovascularization, and which reports, explicitly or implicitly, improvements in corneal ulcers/wound healing [312,313,314,315].
There is no report of angiostatin-induced intraocular pressure increase.
Likewise, there is no report of the application of angiostatin on tendon healing. Interestingly, angiostatin-functionalized collagen scaffolds suppress angiogenesis; histologically, after 8 weeks, the scaffolds with angiostatin exhibited fewer inflammatory cells and more collagen matrix formation [316].

3.1.5. TSP-1, TSP-2

Foulsham et al. (2019), in a comprehensive review, summarizes TSP-1′s roles in ocular surface homeostasis, corneal wound healing, infectious keratitis, and anti-(lymph) angiogenesis; also, it notes the benefit of topical recombinant TSP-1 in mouse models [317]. Classic proof-of-concept that exogenous TSP-1 promotes corneal epithelial closure was adding purified TSP-1 to wounded corneal explants that accelerated re-epithelialization, while anti-TSP-1 antibody inhibited it [318]. In a dry-eye epitheliopathy model, topical recombinant TSP-1 reduced dendritic-cell maturation, inflammatory cytokines, and clinical severity [319]. The effects of TSP1 on hypoxia-induced damages and wound-healing activity in human corneal epithelial (HCE) cells demonstrate that TSP-1-induced exosomal proteins attenuate hypoxia-induced paraptosis in corneal epithelial cells and promote wound healing [320].
Thus, in consideration of evidence of TSP-1 or TSP-2 application alone inducing corneal ulcer healing, existing studies suggest that these proteins play significant roles in regulating angiogenesis and extracellular matrix assembly, processes crucial for corneal wound healing [1,57,58].
There is no direct study showing that exogenous application of TSP-1 or TSP-2 alone (e.g., topical, intracameral, intravitreal, or systemic) directly induced an intraocular pressure rise. However, there is genetic and knockout evidence showing TSPs modulate IOP, and mechanistic/cellular work showing exogenous TSP-1 can reduce outflow facility in perfused anterior segments (which would be consistent with intraocular pressure elevation) [321,322,323,324].
Likewise, there is no direct study showing that exogenous application of TSP-1 or TSP-2—i.e., giving TSP-1 or TSP-2 alone (topically, intratendinous, systemic, etc.)—affects tendon-healing outcome in an in vivo tendon model. However, there is other evidence consistent with a worsening effect on tendon healing. TSP isoforms regulate connective-tissue/tendon structure and healing responses (genetic (knockout/transgenic) studies), and elevated endogenous TSP-2 (e.g., in diabetes) is associated with impaired healing, given that TSP-1/TSP-2 can drive profibrotic/anti-angiogenic/ECM-remodeling pathways that could worsen repair [325,326,327,328,329].

3.1.6. IFN-α

Human case reports/series [330,331,332,333,334] report corneal ulcer healing or remission following IFN-α (systemic or topical) in selected cases (Mooren’s ulcer, refractory herpetic keratitis, HCV-associated peripheral ulceration).
Miller et al. (1993) showed regression of experimental iris neovascularization with systemic IFN-α in primates (strong anti-angiogenic signal in that model) [335], while others showed limited efficacy in the treatment of corneal neovascularization, with subcutaneous IFN-α [336] or subconjunctival (IFNα-2a) [337].
There are clinical reports that document increased intraocular pressure/glaucoma occurring during interferon-α (including peg-IFN α) therapy [338,339,340,341].
Primary manuscripts showed that application of IFN-α alone could clearly neither improve nor worsen tendon healing, had no effect on adhesion or repair strength, or described antifibrotic/anti-contraction effects in vitro [342,343,344,345].

3.2. Summary of the Antiangiogenic Agents in Terms of the “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Furthermore, to implement the cytoprotection concept in eye therapy, in particular in corneal neovascularization counteraction, perceiving BPC 157 corneal ulcer healing capability as a whole was carried out, respecting the antiangiogenic agents, endostatin, PAI-1, PEDF, angiostatin, TSP-1/2, IFN-α, as a class. Commonly, comparing corneal ulcer healing with the “triad” (corneal ulcer healing ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon)↑-relation under a cytoprotection umbrella (e.g., ↑ healing = beneficial, ↓ neovascularization = beneficial, ↑ intraocular pressure = harmful), these antiangiogenic agents revealed the particular different corneal healing/corneal neovascularization/intraocular pressure “Triad” relations, connected or no connected with tendon healing, likely specific for any of these agents’ activities organization (Table 4). However, some caveats should be present given the lack of some direct evidence. Endostatin exhibited particular relations (corneal ulcer healing ↑ remained not determined↔corneal neovascularization ↓ ↔ intraocular pressure not affected (0)↔linked to other avascular tissues healing (i.e., tendon) ↓ (based on the effect on skin lesion). PAI-1 exhibited distinctive relations (corneal ulcer healing ↑ ↔ corneal neovascularization ↓↑ (pro-angiogenic at physiological concentrations and anti-angiogenic at higher concentrations)↔intraocular pressure not affected (0) or ↑ (PAI-1 contributes to glaucoma pathogenesis)↔linked to other avascular tissues healing (i.e., tendon) ↓). PEDF also exhibited distinctive relations (corneal ulcer healing ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↑ ↔ linked to other avascular tissues healing (i.e., tendon) ↑). Angiostatin also evidenced a distinct course (corneal ulcer healing ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure not affected (0)↔linked to other avascular tissues healing (i.e., tendon) not determined (0) or ↑ (given angiostatin-functionalized collagen scaffolds). TSP-1, TSP-2 exhibited a distinct course (corneal ulcer healing ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure not affected (0) or ↑ (genetic studies)↔linked to other avascular tissues healing (i.e., tendon) not determined (0) or ↓ (TSP-1/TSP-2 can drive profibrotic/anti-angiogenic/ECM-remodeling pathways)). IFN-α also evidenced a distinct course (corneal ulcer ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure not affected (0)↔linked to other avascular tissues healing (i.e., tendon) not determined (0)).
Thus, it is commonly acknowledged that endostatin, PAI-1, PEDF, angiostatin, TSP-1/2, and IFN-α should act from the viewpoint of corneal neovascularization, and counteraction, in order to obtain adequate and safe healing of the corneal ulcer [56]. However, with “triad” presentation and connection with tendon healing, as an additional key, this could not be conceptualized. It seems that the various mechanisms and multiple factors, and different standpoints to approach corneal ulcer healing and corneal neovascularization, are all not resolving, providing the extent of perilous events commonly present. On the other hand, whatever the specific mechanism involved, by contrasting different antiangiogenic agents (endostatin, PAI-1, PEDF, angiostatin, TSP-1/2, IFN-α), their divergent cross-tissue effects were highlighted. This helps expose the “fragmentation problem” emphasized earlier.

3.3. Treatment of Corneal Neovascularization in Terms of the “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Treatment of corneal neovascularization is either topical (corticosteroids and non-steroidal anti-inflammatory drugs (NSAIDs), cyclosporine A, anti-VEGF drops) or surgical (laser photocoagulation, diathermy/fine needle cauterization, superficial keratectomy, subconjunctival injections of anti-VEGF drugs, amniotic membrane transplantation, corneal transplantation). However, corneal neovascularization treatment is not always effective and can produce side effects [56].

3.3.1. Corticosteroids

Experimental/clinical data show that topical/systemic corticosteroids—when used alone or before appropriate antimicrobial therapy—can delay or aggravate corneal ulcer/keratitis healing [346,347,348,349,350,351].
Corticosteroids (topical/subconjunctival/systemic) or angiostatic steroid formulations—used alone—inhibit or counteract corneal neovascularization [352,353,354,355,356,357].
Administration of corticosteroids alone (topical, local, systemic, or intraocular) can induce elevation of intraocular pressure (steroid-induced ocular hypertension/steroid glaucoma) [358,359,360,361,362,363,364,365,366].
Administration of corticosteroids alone (local/intratendinous, subacromial, systemic in some models) can impair or aggravate tendon healing, reduce tensile strength, or cause degenerative changes [367,368,369,370,371,372,373,374].

3.3.2. NSAIDs

There is consistent general evidence for the aggravation of corneal ulcer healing due to the application of NSAIDs [375,376,377,378,379,380].
Topical/systemic NSAIDs alone (cyclooxygenase inhibitors) reduced or inhibited corneal neovascularization [381,382,383,384,385].
There is no report that topical/systemic NSAIDs given alone cause clinically meaningful intraocular hypertension. On the other hand, there is strong evidence that NSAIDs antagonize, interfere with, or modify the intraocular pressure-lowering effect of prostaglandin analogues (and in some designs produce small relative increases vs. controls, typically ~1 mmHg) [386,387,388,389,390,391].
Finally, there is evidence that NSAID (COX inhibitor) administration alone aggravated (impaired/delayed or weakened) tendon or tendon-to-bone healing [392,393,394,395,396].

3.3.3. Cyclosporine A

There are many manuscripts (human and animal) reporting that cyclosporine A application—topical or systemic—was associated with corneal ulcer healing [397,398,399,400,401,402].
Likewise, many manuscripts report that cyclosporine A (topical or systemic) application alone counteracted or inhibited corneal neovascularization (animal models and experimental studies) [397,398,399,400,401,402,403,404,405,406,407].
Application of cyclosporine A alone counteracted or reduced increased intraocular pressure (chiefly by substituting cyclosporine A for topical corticosteroids in steroid-induced ocular hypertension/post-keratoplasty glaucoma, and in steroid-intolerant patients) [408,409,410].
There is no report of cyclosporine on tendon healing. However, several mechanistic papers (e.g., [411]) show that cyclosporine A interferes with collagen folding/processing and cultured-cell collagen production; multiple animal wound-healing studies [412,413,414,415] report delayed or impaired healing or altered granulation/collagen deposition. A focused tendon histology study [416] reported no morphologic change in tendons after long-term low-dose cyclosporine A (but did see muscle changes).

3.3.4. Anti-VEGF Drugs

Peer-reviewed experimental manuscripts are showing that anti-VEGF (primarily bevacizumab given subconjunctivally in alkali-burn models) accelerated restoration of the basement membrane, reduced haze, and improved corneal transparency—i.e., promoted corneal wound/ulcer healing [417,418]. Contrarily, several animal studies [419,420,421] and some clinical observations [422] show that topical anti-VEGF (eye-drop formulations, higher concentrations, longer duration) can delay epithelial closure, cause epitheliopathy, or increase stromal remodeling [419,420,421]. Dose, route (subconjunctival vs. topical), timing, model (alkali burn vs. epithelial debridement), and species matter a lot. These healing controversies (promoted vs. impaired) were explained, claiming that anti-VEGF can reduce neovascularization and inflammation (useful), but VEGF also plays roles in normal wound healing, so blocking it may impair epithelial regeneration depending on how it is used [423,424].
In treating corneal neovascularization, studies collectively support the efficacy of topical anti-VEGF therapies, particularly ranibizumab and bevacizumab, in human clinical case series/trials [425,426,427]. There are animal experimental studies demonstrating inhibition [428,429,430,431], and ranibizumab studies [431,432] showing hem- and lymphangiogenesis inhibition.
Evidence of increased (sustained or persistent) intraocular pressure following intravitreal anti-VEGF injections is presented in many reports and case series [433,434,435,436,437,438,439,440].
The evidence that anti-VEGF/tendon healing goes with high VEGF/neoangiogenesis correlates with impaired biomechanical properties [441], and improved tendon-related outcomes after anti-VEGF (VEGF-blocking) intervention (in vivo or clear mechanistic evidence) [442,443,444]. Notably, local injections of an anti-VEGF antibody (B20.4-1-1) given late in the healing course caused a temporary reduction in mechanical properties at an early time point (day 14) when vascularity was reduced—followed by later improvement at a subsequent time point [445].

3.3.5. Summary of Treatment of Corneal Neovascularization in Terms of the “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Furthermore, to implement the cytoprotection concept in eye therapy, in particular in corneal neovascularization counteraction by treatment of corneal neovascularization (corticosteroid, NSAIDs, cyclosporine A, anti-VEGF drops), perceiving BPC 157 corneal ulcer healing capability as a whole was carried out, respecting the mentioned agents used in the treatment of corneal neovascularization as a class. Commonly, comparing corneal ulcer healing with the “triad” (corneal ulcer healing ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↑-relation under a cytoprotection umbrella (e.g., ↑ healing = beneficial, ↓ neovascularization = beneficial, ↑ intraocular pressure = harmful), corticosteroid, NSAIDs, cyclosporine A, and anti-VEGF agents appear to be outside, and revealed no common effect. Contrarily, there are particular different corneal ulcer healing/corneal neovascularization/intraocular pressure “Triad” relations, connected or not connected with tendon healing, likely specific to any of these agents’ activities organization (Table 5). Corticosteroids, with counteraction of corneal neovascularization, exhibited particular detrimental relations (corneal ulcer healing ↓ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↑ ↔ linked to other avascular tissues healing (i.e., tendon) ↓). NSAIDs exhibited a similar chain of events (corneal ulcer healing ↓ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↑ ↔ linked to other avascular tissues healing (i.e., tendon) ↓). On the other hand, cyclosporine and anti-VEGF drugs could be regarded as “fragmented and context-dependent” rather than strictly negative. Cyclosporine A exhibited a different course (corneal ulcer healing ↑ ↔ corneal neovascularization ↓↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↓). Anti-VEGF drops exhibited an additional different course, which is quite complex (corneal ulcer healing ↑ (reduced neovascularization and inflammation) ↓ (inhibition of normal healing (VEGF blocked))↔corneal neovascularization ↓↔ intraocular pressure ↑ ↔ linked to other avascular tissues healing (i.e., tendon ↓ (early points), ↑ (late points)) (Table 5).
Thus, with respect to corneal ulcer healing/corneal neovascularization (↓ neovascularization ≈ ↑ corneal ulcer healing), corticosteroid and NSAIDs act differently, and depart to opposite relation (↓ corneal neovascularization ≠ ↑ corneal ulcer healing). There is a delayed corneal ulcer healing along with counteracted corneal neovascularization. Likewise, the effects of cyclosporine A, and anti-VEGF agents clearly preclude the suggested mechanisms as a way to achieve safe corneal ulcer healing. Furthermore, with a full “triad” presentation (↑ intraocular pressure (corticosteroid, NSAIDs, anti-VEGF agents), ↓ intraocular pressure (cyclosporine)), and connection with tendon healing (quite consistently impaired tendon healing (except for the later interval with anti-VEGF), as an additional key, this could not be conceptualized, and divergent cross-tissue effects were highlighted.
On the other hand, as mentioned before, these favorable points (↓ neovascularization ≈ ↑ corneal ulcer healing) may be more supported by BPC 157 therapy for corneal ulcer healing [3]. Besides being combined with maintaining transparency and counteracting corneal lesions after different injuries [3,38,39,40], but favoring an angiogenesis healing effect in different tissue healing [4], in particular in those avascular, like tendon and ligament [42,43,44,45,46,47,48,49], as emphasized before, there is additional evidence of an orchestrated effect, depending on the tissue, time, and injury [4]. Notably, recently, cornea healing “angiogenic privilege” was reviewed as a part of BPC 157 pleiotropic effects [4]. It appears as a therapy and safety key, targeting angiogenesis and NO’s cytotoxic and damaging actions, but maintaining, promoting, or recovering their essential protective functions, a special beneficial effect controlling and modulating angiogenesis and the NO-system, thus, along with advanced tendon and other tissues healing as a proof of the concept [4]. Consistent with such a modulatory and controlling role, BPC 157 therapy strongly counteracted the adverse effects of both corticosteroids [4,10,43,45] and NSAIDs [4,10,446]. Moreover, consistent with such a role, BPC 157 retrobulbar application directly mitigated the detrimental retrobulbar application of NO-blocker, L-NAME in rats, and L-NAME-induced retinal ischemia [37]. Fundoscopy demonstrated strong generalized irregularity of the diameter of blood vessels with severe atrophy of the optic nerve, and extremely poor presentation of the choroidal blood vessels. Consistently, there was a progressively worsened course, degeneration of ganglion cells, and narrowing of vascular lumina in the nerve cell layer (Factor VIII immunohistochemistry), marked damage in the inner plexiform and inner nuclear layers, accompanied by reduced retinal thickness and evidence of complete retinal injury. BPC 157 therapy may induce a prompt recovery, either given 20 min after L-NAME or 48 h after L-NAME, and markedly improved animal behavior on days and weeks after retrobulbar application of L-NAME [37]. Unlike growth factors [447], consistent with Folkman’s key concept (inhibited corneal neovascularization, inhibited tumor growth) [448,449,450,451], in the human melanoma cell line, BPC 157 inhibits the VEGF effect [73], attributed to controlling the VEGF system, as well [4,49,70,71,72]. Furthermore, in mice with C26 colon adenocarcinoma, BPC 157 counteracted tumor cachexia and severe muscle wasting, corrected deranged muscle proliferation and myogenesis, counteracted weight loss, and markedly prolonged survival [79].

4. Treatment of Glaucoma in Terms of the “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Recently, it was pointed out that such a wide cytoprotection agenda in glaucoma therapy might also be distinctive from the focused background of the alpha 2-agonists, beta-blockers, inhibitors of carbonic anhydrase, or parasympathomimetics, ROCK/Rho-kinase (Rho) inhibitors, and prostaglandin derivatives [9].

4.1. Alpha 2-Agonists

Randomized controlled trial data on corneal ulcer healing due solely to alpha 2-agonists used in glaucoma therapy are not available. On the other hand, many manuscripts, case reports, case series, and an in vitro mechanistic study document adverse corneal effects from topical alpha 2-agonist antiglaucoma agents (i.e., brimonidine and multi-drug regimes including brimonidine)—evidence that these agents can aggravate or complicate corneal disease/healing, and can cause corneal epithelial/stromal inflammation, sterile infiltrates, pseudodendritic lesions, neovascularization, and persistent stromal opacity [452,453,454,455,456,457,458].
For both commonly used topical alpha-2 agonists (brimonidine, apraclonidine), the peer-reviewed clinical literature consistently documents that intraocular pressure lowering begins within about 1 h and peaks at ~2–3 h after a topical dose [459,460,461,462,463,464].
Considering the possible effect on tendon healing, although there is no study of the direct effect on tendon injury of alpha 2-adrenergic agonists used in anti-glaucomatous therapy, alpha 2A-adrenoceptors have been detected in ocular tissues (ciliary epithelium, trabecular meshwork, retina) and in extraocular tissues (e.g., tendons, skin fibroblasts). Stimulation may affect cell proliferation, vascular tone, and inflammation [465,466,467,468,469,470]. Like for corneal pathology, the overall weight of evidence indicates that alpha 2-agonist activity is detrimental (negative effect) for tendon healing. Specifically, direct experimental alpha 2-agonist stimulation in tendon induces tendinosis-like hypercellularity [471]. Human biopsy/immunohistochemistry studies [472,473] suggest adrenergic signaling sustains tendinopathy pathology. Additionally, a systematic review linked sympathetic/adrenergic activity with tendinopathy [474]. Cutaneous wound healing in a knockout mouse model demonstrated that alpha 2 receptor signaling slows repair; absence improves healing [470].

4.2. Beta-Blockers

Beta-blockers used for glaucoma (timolol or related beta-adrenergic antagonists) accelerated or improved corneal epithelial wound/ulcer healing [475,476,477].
Likewise, there is direct evidence that beta-blocker agents used in glaucoma therapy (timolol, propranolol, etc.) counteract corneal neovascularization [478,479,480]. Contrarily, far less consistent appear to be the negative or mixed reports where beta-blockers (timolol, propranolol, betaxolol) were tested against corneal neovascularization but did not consistently inhibit or sometimes had no effect, or were less robust than the comparative agent [481,482,483].
Topical beta-blockers (e.g., timolol) begin to lower intraocular pressure within ~15–30 min after instillation; the maximal intraocular pressure reduction is usually reached within ~1–3 h, and the effect commonly persists ~12–24 h with single doses [484,485,486].
Considering the possible effect on tendon healing, there is no study of the direct effect on tendon injury of beta-blockade used in anti-glaucomatous therapy. However, there are beneficial effects of beta-blockade on inflammation, re-epithelialization, angiogenesis, and bone formation—mechanisms that overlap with phases of tendon repair (inflammation modulation, matrix remodeling, vascular responses). The animal and human burn/fracture clinical data [487,488,489,490] are especially load-bearing for systemic beta-blocker effects on repair. The topical timolol case series and small randomized scar studies [491,492,493] show local beta-blockade can promote epithelial/dermal repair when applied to skin wounds.

4.3. Carbonic Anhydrase Inhibitors

No rigorously controlled, PubMed-indexed studies have directly demonstrated that topical or systemic carbonic anhydrase inhibitors negatively affect corneal ulcer epithelial re-epithelialization as a primary outcome. However, there is a consistent set of PubMed-indexed clinical reports, case series, and clinical studies showing that topical carbonic anhydrase inhibitors (dorzolamide, brinzolamide ± combinations) can cause or worsen corneal edema/endothelial decompensation or surface epithelial damage in eyes with compromised corneal endothelium or ocular surface. These effects would reasonably aggravate healing of a corneal ulcer in those vulnerable eyes. Multiple case series and case reports document corneal edema/endothelial decompensation temporally associated with the use of topical carbonic anhydrase inhibitors (dorzolamide, brinzolamide), sometimes reversible on cessation and in some reports irreversible—clearly relevant because corneal edema and endothelial failure worsen outcomes and delay epithelial healing of ulcers [494,495,496]. Randomized/controlled and short-term studies show measurable increases in central corneal thickness or changes in corneal hydration control in patients with endothelial compromise or cornea guttata after dorzolamide, i.e., a physiologic basis for impaired corneal deturgescence that would impede ulcer healing [497,498]. Clinical reviews/practice notes advise caution or prefer systemic carbonic anhydrase inhibitors in the setting of infected or compromised corneas because topical carbonic anhydrase inhibitors may be toxic to damaged endothelium or ocular surface—practical guidance consistent with the clinical reports above [499].
No PubMed-indexed controlled trials have proven that carbonic anhydrase inhibitors directly cause corneal neovascularization. However, there are a small number of PubMed-indexed case reports/case series and one narrative/case-report review in which corneal stromal sterile infiltration with neovascularization occurred in patients who were using topical antiglaucoma medications that included a carbonic anhydrase inhibitor (brinzolamide or dorzolamide)—typically in the context of multiple topical agents [500,501,502].
Intraocular pressure reduction with topical carbonic anhydrase inhibitors (dorzolamide, brinzolamide) starts within ~1 h. Intraocular pressure reduction with systemic carbonic anhydrase inhibitors (acetazolamide, methazolamide) starts within 1–2 h (acetazolamide) and 2–4 h (methazolamide) [503].
There are no PubMed-indexed animal experiments or clinical studies in which topical or systemic carbonic anhydrase inhibitors used at glaucoma doses were shown to aggravate or improve tendon healing. However, certain carbonic anhydrase isoforms are upregulated during skin wound healing, and the activity of carbonic anhydrases can influence re-epithelialization—useful background for carbonic anhydrase roles in tissue repair [504]. More specifically, carbonic anhydrases (CAs) are involved in many pathological conditions, and the overexpression of both CA9 and 12 in inflamed joints has been recently reported. Consequently, a selective CA9/12 inhibition could be a feasible strategy for improving tendon recovery after injury, evidenced in an in vitro study on human tenocytes showing carbonic anhydrase-targeting compounds can change inflammatory signaling in tendon cells [505].

4.4. Muscarinic Agents (Including Pilocarpine)

In controlled animal studies, muscarinic agents (including pilocarpine) used in anti-glaucomatous therapy improve corneal epithelial defect/ulcer healing [506,507]. Contrarily, there is a lack of effect of 2% pilocarpine on corneal epithelial healing in rabbits [508]. Moreover, an in vitro study on primary human corneal stromal cells found dose- and time-dependent cytotoxicity of pilocarpine (apoptosis, membrane permeability, DNA fragmentation, caspase activation) at concentrations above ~0.625 g/L [509]. Likewise, using rabbit corneal endothelium, in vitro perfusion, and electron microscopy, there is a dose-related endothelial toxicity (cell shrinkage, nuclear margination, cytoplasmic vacuolation). This is the basis for statements that pilocarpine (especially at higher concentrations/exposure) can harm corneal endothelium in animal models [510]. These results are consistent with other studies [511,512]. Additionally, there is a reported pilocarpine-associated allograft rejection in post-keratoplasty patients [513]. On the other hand, measuring corneal epithelial healing and acetylcholine content in the corneal epithelium found that higher acetylcholine levels were associated with faster healing, thus, a positive correlation between endogenous acetylcholine level and wound healing [514].
Consequently, as a result, no studies directly demonstrate that muscarinic agents used in glaucoma treatment either inhibit or promote corneal neovascularization. Still, the mentioned Massry & Assil (1995) study in a clinical case series (three cases) described a temporal association between initiation of topical pilocarpine and corneal graft rejection—inflammatory events that can be accompanied by neovascularization in some contexts [513].
Considering the lowering of the increased intraocular pressure, pilocarpine acts within minutes to an hour and lasting several hours (formulation dependent). Short-acting acetylcholinesterase inhibitors (physostigmine) show rapid daytime effects (onset in tens of minutes and peak within 1–2 h). Echothiophate produces miosis within an hour but exhibits a delayed peak intraocular pressure decrease (~24 h) and very prolonged duration (days–weeks). Carbachol intracameral application acts essentially immediately (minutes) in the intraoperative setting with topical preparations lowering intraocular pressure over the first hours [514,515,516,517,518].
No direct manuscripts show that muscarinic agents (pilocarpine, carbachol, or other) used in glaucoma therapy either counteracted or improved tendon healing. However, although without testing pilocarpine or other glaucoma-used muscarinic drugs as tendon therapeutics, some papers provide related mechanistic/preclinical evidence (acetylcholine/acetylcholinesterase-inhibitor). They show cholinergic stimulation (exogenous acetylcholine or increasing acetylcholine by inhibiting acetylcholinesterase), and that muscarinic receptor activation in tenocytes can enhance tenocyte proliferation or improve bone–tendon (enthesis) healing in animal models [519,520]. Therefore, muscarinic signaling may facilitate tenocyte proliferation through a cellular mechanism.

4.5. Rho-Kinase Inhibitors

Rho-kinase inhibitors are (or are the same class as) drugs used in glaucoma therapy (ripasudil, netarsudil), which can improve corneal wound/ulcer or epithelial/endothelial healing [521,522,523,524].
The evidence about Rho-kinase inhibitors and corneal neovascularization suggests the reduction in corneal neovascularization. Of the ROCK inhibitors that are actually used clinically for glaucoma, ripasudil (K-115) has clear, peer-reviewed evidence showing a reduction in corneal neovascularization in an experimental corneal transplant model (see Inomata et al.) [525]. Several other ROCK inhibitors (notably fasudil and the research compound Y-27632) have preclinical evidence for anti-angiogenic effects (in vitro or in animal CNV/retinal models)—these support the mechanism but are not (in most countries) glaucoma drugs in routine clinical use [526]. For netarsudil (an FDA-approved glaucoma drug), the evidence includes corneal hemorrhage or corneal vascularization temporally associated with netarsudil use—so the clinical/real-world corneal effects of netarsudil are mixed and may even include adverse vascular events in some settings [527].
Illustrative is a Phase 2 clinical trial (0.4% ripasudil in primary open angle glaucoma/ocular hypertension therapy patients), where peak intraocular pressure lowering was observed 2 h after instillation, while a trough (lowest effect before the next dose) was recorded just before the next instillation [528].
There are no manuscripts showing that the Rho/ROCK inhibitors that are used clinically for glaucoma (i.e., topical ripasudil or netarsudil) improve tendon healing. The glaucoma-drug literature is focused on ocular tissues (trabecular meshwork, cornea, Tenon’s fibroblasts, etc.), not musculoskeletal tendon repair. Mechanistic, preclinical evidence exists: multiple lab studies show that inhibiting ROCK (commonly with Y-27632 or related inhibitors) affects tendon stem/progenitor cell mechanics and can enhance tenogenic differentiation in vitro. Those findings suggest a mechanistic plausibility that ROCK modulation could help tendon repair, but they do not prove that the clinical glaucoma drugs (ripasudil/netarsudil) will do so in vivo [529,530].

4.6. Latanoprost

Latanoprost, in terms of corneal ulcer healing, exhibited a dual effect. It improved corneal ulcer healing [531,532,533,534,535,536] consistent with commonly acknowledged evidence that just preservative-free or alternative-preservative prostaglandin formulations (or cationic-emulsion latanoprost) have reduced corneal toxicity and better wound-healing profiles. Contrarily, latanoprost (especially benzalkonium chloride (BAK)-preserved formulations) can aggravate corneal surface injury or delay corneal wound healing/recur herpetic keratitis [531,535,537,538,539,540].
Latanoprost, in terms of corneal neovascularization, exhibited a dual effect. Prostaglandin analogues can promote corneal angiogenesis [539,540,541,542,543,544]. Illustrative as direct evidence that prostaglandin analogues can promote corneal angiogenesis in certain models is the study in a rat corneal micropocket model [545], where prostaglandin formulations produced the most prominent angiogenic stimulatory effect among several antiglaucoma drugs tested. Consistently, as mentioned, in a case report of stromal corneal neovascularization temporally associated with latanoprost therapy, corneal neovascularization regressed after discontinuation—which underscores that in real patients latanoprost/its formulation can be linked to corneal neovascularization [541]. On the other hand, preservative-free/cationic-emulsion latanoprost formulations have markedly better ocular-surface tolerance and promote corneal epithelial healing and reduced surface inflammation compared with BAK-preserved latanoprost, which may reduce the risk of secondary corneal neovascularization as an indirect effect [531,532,533,536].
Measurable intraocular pressure reduction begins within a few hours after topical dosing. Clinical single-dose studies report the average time to 50% of maximal intraocular pressure reduction (a practical “onset” measure) of about 6.0 h [546].
No direct evidence currently links topical latanoprost to affect tendon healing. However, a dual role could be suggested. This was based on evidence that local PGE2 application increased some structural strength metrics, illustrating that prostaglandin signaling modulates tendon remodeling and mechanics [547]. On the other hand, an in vitro study demonstrated that low levels of PGE2 can support tendon remodeling and healing, while high levels of PGE2 impair tendon stem cell proliferation and push them toward non-tenocyte lineages (bone, fat, cartilage), which is detrimental to tendon repair [548].

4.7. Summary of Treatment of Glaucoma in Terms of the “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Furthermore, to implement the cytoprotection concept in eye therapy, in particular in the treatment of glaucoma (alpha 2-agonists, beta-blockers, carbonic anhydrase inhibitors, muscarinic agonists (including pilocarpine), Rho-kinase inhibitors, latanoprost), perceiving BPC 157 corneal ulcer healing capability as a whole was carried out, respecting the mentioned agents used in the treatment of glaucoma as a class. Commonly, comparing corneal ulcer healing with the “triad” (corneal ulcer healing ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↑-relation under a cytoprotection umbrella (e.g., ↑ healing = beneficial, ↓ neovascularization = beneficial, ↑ intraocular pressure = harmful), most of agents appear to be outside, while beta-blockers and probably Rho-kinase inhibitors appear to be inside, and revealed no common effect. This can be seen with the reduction of aqueous production (alpha 2-agonists, beta-blockers, carbonic anhydrase inhibitors) as well as with increases of uveoscleral outflow of aqueous humor (muscarinic agonists (including pilocarpine), Rho-kinase inhibitors, latanoprost). Thus, with the caveat that some direct evidence is lacking, there are particular different corneal ulcer healing/corneal neovascularization/intraocular pressure “Triad” relations, connected or not connected with tendon healing, likely specific for any of these agents’ activities organization (Table 6). Alpha 2- agonist, with counteraction of increased intraocular pressure, exhibited particular detrimental relations (corneal ulcer healing ↓ ↔ corneal neovascularization ↑ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↓). Carbonic anhydrase inhibitors exhibited a distinct chain of events (corneal ulcer healing ↓ ↔ corneal neovascularization ↑ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↑ (effect supposed as carbonic anhydrases are involved in many pathological conditions). On the other hand, β-blockers’ counteraction of increased intraocular pressure exhibited a promising course (corneal ulcer healing ↑ ↔ corneal neovascularization ↓ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↑ (based on the effect on skin lesion)). Muscarinic agents (including pilocarpine) provide a counteraction of increased intraocular pressure along with a particular course (i.e., dual effect on corneal ulcer healing) (corneal ulcer healing ↑ ↓ ↔ corneal neovascularization ↑ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↑ (enhanced tenocyte proliferation). Rho-kinase inhibitors exhibited a counteraction of increased intraocular pressure along with a course similar to that of beta-blockers (corneal ulcer healing ↑ ↔ corneal neovascularization (corneal ulcer healing ↑ ↔ corneal neovascularization, ripasudil, fasudil ↓, netarsudil ↑ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon) ↑ (tendon stem/progenitor cell mechanics). Latanaprost-induced lowering of intraocular pressure is frequently accompanied by other dual effects, and thereby, there is a course that cannot be easily estimated (corneal ulcer healing, preservative-free latanoprost ↑, BAK-preserved products ↓ ↔ corneal neovascularization preservative-free latanoprost ↓, BAK-preserved products ↑ ↔ intraocular pressure ↓ ↔ linked to other avascular tissues healing (i.e., tendon, low level of PGE2 ↑, high level of PGE2 ↓).
Notably, for current glaucoma therapies, although all current glaucoma therapies are acknowledged as potent and effective intraocular pressure-lowering agents, they require substantial time to start and exert their lowering effects and do not share a unifying therapeutic concept. Moreover, combined with the other avascular tissue healing (i.e., tendon), the standard agents used in glaucoma therapy lack the common concept even more, given the corneal ulcer healing/corneal neovascularization/intraocular pressure “triad”. Furthermore, almost any one of the particular concepts, presenting very distinctive drug classes, shows particular discrepancies and limited effectiveness (Table 6). Thereby, for the success of glaucoma therapy, as intraocular pressure-lowering agents, the additional combining of these principles can be indicative since very early times (i.e., pilocarpine and epinephrine [549,550,551], timolol concomitant to epinephrine to enhance the ocular hypotensive effects in many patients [552]). With respect to corneal ulcer healing/corneal neovascularization/intraocular pressure, the “triad”, combined with the other avascular tissue healing (i.e., tendon), the indicated encouraging effect of timolol could be confounded with beta-blockers started with propranolol. Pitfalls encountered were intravenous applications lowering intraocular pressure, avoiding corneal anesthetic properties, which have produced negative effects on tear production, profound dry eye syndrome, subconjunctival fibrosis, and tachyphylaxis [553]. Finally, there is the ambiguous corneal ulcer healing/corneal neovascularization/intraocular pressure, the “triad”, combined with the other avascular tissue healing (i.e., tendon) noted with latanoprost, a resolving prostaglandin analogue. Therefore, the proposed selective activation of the FP receptor in ciliary muscle and adjacent tissues, remodeling of the extracellular matrix, and upregulating matrix metalloproteinases to increase uveoscleral outflow of aqueous humor [41], does not correlate with the proposed cytoprotection context once related to prostaglandins’ cytoprotective role [13,14,15,16,17,18,19,20,21], while sustained reduction in intraocular pressure requires more hours.
Thus, there is a new and more complex point with BPC 157 therapy in glaucomatous rats, and eye therapy, in general [3,9,35,36,37,38,39,40,42,43,44,45,46,47,48,49]. Presenting quite an immediate lowering effect on the increased intraocular pressure, and with respect to the known therapy, achievement, and pitfalls [549,550,551], the proposed BPC 157 cytoprotective therapy can be an additional resolving principle [4,5,6,7,8,9,10,11,12,23,24,25,26]. Notably, it covers the recovery of glaucomatous rats [9,35] and normalized intraocular pressure [35], maintenance of retinal integrity [35,37], recovery of pupil function [36,37], recovery of retinal ischemia [37], counteracting of corneal neovascularization [35], and corneal injuries [3,38,39,40]. In particular, ascertained transparency maintenance [3,38,39,40], counteracting corneal drying, counteracting the loss of corneal sensation, counteracting the decrease in blink rate, and maintaining tear production [39,40] have equal effectiveness for topical and systemic application. The specificity of the therapy [4] of BPC 157 that should be requested is confirmed also in several tendon injury models [42,43,44,45,46,47,48,49].
Thereby, the proposed BPC 157 cytoprotective principle should be more viable in glaucomatous rats. Three episcleral vein cauterizations, and intraocular pressure just exceeding 30 mmHg, rapidly counteracted BPC 157 therapy, which underlines the cytoprotective ability of the pentadecapeptide BPC 157 to further maintain and upgrade endothelium integrity and functioning (for review see [4]), particularly focused on minor vessels during noxious procedures, rapidly upgrading function to substitute the failed major blood vessels. Illustratively, this likely recovered glaucomatous rats, where one episcleral vein remained, and took over the failed function of all the episcleral veins, amid rapid lowering of increased intraocular pressure and subsequent retinal integrity preservation, as proof [9,35]. These findings show, along with a systemic beneficial effect, the recovery of the severe occlusion/occlusion-like syndrome [554,555,556,557,558,559,560,561,562,563,564,565,566,567,568,569,570,571]. There, the prompt activation of collateral pathways, “bypassing vascular key”, i.e., activated azygos vein to direct blood flow delivery, results in the recovery of multiorgan failure syndrome. This occurred in the rats with vascular failure induced by major vessels’ occlusion, peripherally [554,555,556,557,558,559] and centrally [560,561], and other similar noxious procedures [562,563,564,565,566,567,568,569,570,571] that largely affect endothelium function, and multicausal pathology was fully recovered [554,555,556,557,558,559,560,561,562,563,564,565,566,567,568,569,570,571]. There were general blood pressure disturbances (i.e., intracranial (superior sagittal sinus), portal and caval hypertension, and aortal hypotension). The lesions occurred in the brain (including intracerebral and intraventricular hemorrhage), heart (severe arrhythmias, congestion, and endocardial infarction), and lungs (hemorrhage), with congestion in the liver, kidney, and gastrointestinal lesions. Major vessels were congested (i.e., inferior caval vein, superior mesenteric vein), the azygos vein collapsed, venous and arterial thrombosis progressed, peripherally and centrally, and the advanced Virchow triad was fully substantiated. As emphasized, also in the context of the effects noted in BPC 157 eye therapy, these were all attenuated/eliminated by BPC 157 therapy [554,555,556,557,558,559,560,561,562,563,564,565,566,567,568,569,570,571]. Illustratively, major vessel congestion was reversed to normal vessel presentation, the recovered azygos vein reactivated the pathway for direct blood flow delivery, and the vascular failure (and Virchow triad circumstances) was effectively cured [554,555,556,557,558,559,560,561,562,563,564,565,566,567,568,569,570,571]. Furthermore, Fourier transform infrared spectroscopy revealed a rapid change in the lipid content and protein secondary structure conformation within the vessel wall, occurring within minutes, following BPC 157 therapy [572]. This shows support for the vessel’s function even in the worst circumstances. Thus, the BPC 157 beneficial effect of the corneal healing/corneal neovascularization/intraocular pressure “triad” combined with avascular tissue (i.e., tendon) healing occurred as part of its general cytoprotective effect [4].
Finally, we should conclude with resolving transparency, as a particular part of the corneal ulcer healing, and corneal ulcer healing as a part of BPC 157 wound healing therapy [3,4,22,23,24,25]. Notably, inducing simultaneous healing of different tissues specifically depends on the tissue involved, and this should be, in general, along with the cornea healing as a prime point [3,4,22,23,24,25], as a relatively unique tissue in the body, with transparency and a lack of blood vessels [573]. Thus, possessing a somewhat unique defense system [1,2], there is fibrin deposition, a “translucent fibrin clot” in the cornea as an initial scaffold for healing [573]. There, in analogy with specifically demonstrated counteraction of adhesion formation, both prevention and reversal [22,23,24,25], BPC 157 therapy would improve the beneficial course by optimizing clot resolution along with corneal ulcer healing and further improve corneal transparency [3]. Basically, the peritoneum is also unique in its morphology [574] (hemostasis is not relevant [574], as in cornea ulcer healing as well [573]); with damage to the peritoneum, fibrin is deposited. Consequent to fibrin deposition, adhesion formation increases as a sign of necrotic tissue, inflammation, and initial clotting not rapidly removed in a regular course [574,575,576,577,578]. Or, they could be promptly reversed (i.e., by BPC 157 therapy [24,25,575]), as a highlight of an advanced healing course, given that the fibrin matrix is lysed, and normal peritoneal fibrinolytic activity is reestablished [574,575,576,577,578]. Thus, the peritoneum–cornea analogy may serve as an analogous remodeling of fibrin-based repair systems [573,574,575,576,577,578]. Consequently, there is indicative evidence that BPC 157 strongly counteracted adhesion formation following peritoneal excision, and both prevented and reversed those already formed. Likewise, BPC 157 therapy counteracted adhesions along with improved anastomosis healing, and fistula closing, as an effect related to an interaction with the NO-system [24,25,575]. Together, this confirms how BPC 157 may particularly preserve corneal transparency while promoting ulcer healing [3]. In addition, BPC 157 attenuates prolonged bleeding and thrombocytopenia after amputation and anticoagulant or aspirin use as an interaction with the NO-system [83,579], while considering arterial and venous thrombosis counteraction [580], it counteracts the whole Virchow triad as shown in the recovery of the huge pathology in all occlusion/occlusion-like syndromes [554,555,556,557,558,559,560,561,562,563,564,565,566,567,568,569,570,571], mentioned before. Interestingly, when given with aspirin, clopidogrel, or cilostazol, BPC 157 largely rescues thrombocyte function in rats and does not affect coagulation factors [581]. This may be important given that in the cornea, the fibrinolytic system not only participates in the regulation and execution of ECM turnover but also contributes to the control of other physiological functions of the various corneal cell types [573].

5. Summary of Corneal Ulcer Therapy, Angiostatic Factors, Neovascularization Therapy, and Glaucoma Therapy in Terms of the “Triad” Approach (Corneal Ulcer Healing↔Corneal Neovascularization↔Intraocular Pressure)

Quite extensive beneficial effects of BPC 157 therapy, as a cytoprotective agent, in eye therapy [3,9,35,36,37,38,39,40], and also in other avascular tissue, tendon healing [42,43,44,45,46,47,48,49], quite extensive even in the cytoprotection terms, apparently interconnected since obtained with the same dose range [3,9,35,36,37,38,39,40,42,43,44,45,46,47,48,49], accord corneal ulcer healing/corneal neovascularization/intraocular pressure relations, as commonly known mutual interrelation [1,2,29,30,31,32,33,34]. Combined tendon healing provides an avascular tissue benchmark that clarifies an agent’s capacity to restore homeostasis in corneal ulcer disease [4]. This enables more mechanism-driven translation, implemented as a “triad”, where cytoprotection serves as the unifying principle that conceptually links corneal ulcer healing, corneal neovascularization, and intraocular pressure regulation, underscoring that therapeutic effects are not isolated but interconnected. Preclinical studies with BPC 157 therapy, as a cytoprotection agent, illustrate this integration (i.e., the corneal ulcer healing (e.g., ↑)↔corneal neovascularization (e.g., ↓)↔intraocular pressure (e.g., ↓) relation is integrated in one agent’s effect). The extent of integration that could be achieved was investigated by mapping standard therapeutic agents used for corneal ulcers, neovascularization, or glaucoma onto this “triad” and linking them with tendon healing.
This new approach challenges the standard therapies, which are typically designed to address single targets, with the standard agents’ background intended to act in “fragments”. This raises the possibility that these fragmented approaches could be reversed conceptually toward the corneal ulcer healing/corneal neovascularization/intraocular pressure “triad”, and other avascular tissue healing, within an integrated cytoprotection framework. As emphasized before, mapping was conducted despite the caveats due to the absence of some direct evidence, heterogeneity of the models and endpoints, and the majority of mapping being preclinical. Included were the ascorbate, fibronectin, hyaluronic acid, metalloproteinase inhibitors, EGF, FGF, NGF, insulin, and IGF-1 (corneal ulcer healing, Section 2.9 (Table 3)), the antiangiogenic agents (endostatin, PAI-1, PEDF, angiostatin, TSP-1, TSP-2, IFN-α (Section 3.2 (Table 4)), corticosteroids, NSAIDs, cyclosporine A, anti-VEGF drops (treatment of corneal neovascularization (Section 3.3.5 (Table 5)), and alpha-agonists, beta-blockers, carboanhydrase inhibitors, muscarinic agonists, Rho-kinase inhibitors, and prostaglandin analogues (glaucoma, Section 4.7, Table 6). To better illustrate the cytoprotection triad, all of the agents were grouped according to their shared phenotypic profiles (Table 7). Consequently, particular relations between the agents appeared, proving that within corneal ulcer healing/corneal neovascularization/intraocular pressure “triad” relations combined with tendon healing as a prime benchmark of orchestrated avascular tissue healing (i.e., resolved cornea’s “angiogenic privilege”), BPC 157, ascorbate, and insulin appear to uniquely fulfill the entire triad and tendon healing without trade-offs (Table 7). Latanoprost, due to its consistent dual effects (except for consistently lowering intraocular pressure), could not be accommodated. Notably, however, for the cytoprotection background, BPC 157 strongly counteracted the adverse effects of insulin overdose application [582]. This suggests a broad cytoprotective buffering potential, given that BPC 157 is very safe (in toxicology (including ocular-specific safety) studies LD1 not achieved [4,583], no adverse effects in clinical trials [62,63,64,65,584]).
Furthermore, it remains to see the significance of the evidence that beta-blockers and Rho-kinase inhibitors closely follow (although with narrower evidence bases) as well as the significance of NGF as primarily reparative, neutral for corneal neovascularization and intraocular pressure. Together, these may be separate pathways to realize the integrative cytoprotection effect. Latanoprost remains a special issue. The other agents seem to be more (providing some adverse outcome as a shared point) or less (providing some beneficial effects combined, i.e., corneal ulcer healing/tendon healing, despite other adverse outcomes) outside the cytoprotection “umbrella”. Hyaluronic acid and PEDF support tendon/corneal healing but may raise intraocular pressure. Cyclosporine A and MMP inhibitors support corneal healing but impair tendon repair. Fibronectin, EGF, FGF, and IGF-1 promote corneal healing and tendon but simultaneously drive corneal neovascularization and intraocular pressure elevation. Classical anti-angiogenics (endostatin, PAI-1, TSP-1/2, IFN-α) suppress corneal neovascularization but impair repair. Possibly, overlapping effects, shared phenotypic patterns, given that corneal healing/corneal neovascularization/intraocular pressure, combined with tendon healing, should be specific for each of the agents, may suggest shared negative points toward the negative outcomes (i.e., increased intraocular pressure, corneal neovascularization, failed corneal ulcer healing, etc.). On the other hand, summarizing the shared phenotypic patterns can immediately provide an insight into which agents fully, partially, or poorly align with cytoprotection. Finally, to avoid a complex discussion about a multitude of factors involved, for instance, in corneal ulcer pathology, the agents’ effects speak for themselves, even without speaking. Thus, the agents’ profiles, when summarized in this triad framework, clearly reveal their alignment—or misalignment—with cytoprotection, and consequently, their therapy potential.

6. Conclusions

In conclusion, to better resolve corneal ulcer healing, we introduced a novel “triad” (corneal ulcer healing↔corneal neovascularization↔intraocular pressure), as a conceptual framework in ocular therapy, and extended it to avascular tissues such as tendon and cytoprotection; as the shared conceptual anchor, therapeutic effects are not isolated but interconnected. Within this framework, corneal ulcer healing↔corneal neovascularization↔intraocular pressure “triad” properly interrelated, BPC 157 therapy, as a cytoprotection agent therapy, has shown in preclinical studies positively interconnected beneficial effects. There is the ability to rapidly normalize intraocular pressure in glaucomatous rats, preserve retinal integrity, restore pupil function, promote transparent corneal healing, and counteract both corneal neovascularization and dry eye. Likewise, cornea healing “angiogenic privilege” along with advanced tendon healing as proof of the healing concept, as consistent effects, are documented in tendon injury models, underscoring its cytoprotective specificity. This novel approach was substantiated by mapping standard therapeutic agents specifically used for corneal ulcer healing, or neovascularization therapy, or glaucoma onto this triad and linking them to tendon healing. The mapping of different therapeutic domains under one unifying triad reveals for each class a shared framework of different “triad” presentations, likely specific to the agent’s effect, connection, or no connection with other avascular tissue healing. This would unify disparate therapeutic observations but also highlight gaps and inconsistencies across existing drug classes. These findings encourage further translational research into the cytoprotection concept, cytoprotection as the unifying principle, and BPC 157, in particular, as the first exemplar, potential clinical application.

Funding

This work was supported by the University of Zagreb, Zagreb, Croatia (Grant BM 10106-22-3071).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ljubimov, A.V.; Saghizadeh, M. Progress in corneal wound healing. Prog. Retin. Eye Res. 2015, 49, 17–45. [Google Scholar] [CrossRef] [PubMed]
  2. Sharif, Z.; Sharif, W. Corneal neovascularization: Updates on pathophysiology, investigations & management. Rom. J. Ophthalmol. 2019, 63, 119–127. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Masnec, S.; Kokot, A.; Zlatar, M.; Kalauz, M.; Kunjko, K.; Radić, B.; Kliček, R.; Drmić, D.; Lazić, R.; Brcic, L.; et al. Perforating corneal injury in rat and pentadecapeptide BPC 157. Exp. Eye Res. 2015, 136, 9–15. [Google Scholar] [CrossRef] [PubMed]
  4. Sikiric, P.; Seiwerth, S.; Skrtic, A.; Staresinic, M.; Strbe, S.; Vuksic, A.; Sikiric, S.; Bekic, D.; Soldo, D.; Grizelj, B.; et al. Stable gastric pentadecapeptide BPC 157 as a therapy and safety key: A special beneficial pleiotropic effect controlling and modulating angiogenesis and the NO-system. Pharmaceuticals 2025, 18, 928. [Google Scholar] [CrossRef] [PubMed]
  5. Sikiric, P.; Seiwerth, S.; Skrtic, A.; Staresinic, M.; Strbe, S.; Vuksic, A.; Sikiric, S.; Bekic, D.; Penovic, T.; Drazenovic, D.; et al. Acute compartment syndrome and intra-abdominal hypertension, decompression, current pharmacotherapy, and stable gastric pentadecapeptide BPC 157 solution. Pharmaceuticals 2025, 18, 866. [Google Scholar] [CrossRef] [PubMed]
  6. Sikiric, P.; Sever, M.; Krezic, I.; Vranes, H.; Kalogjera, L.; Smoday, I.M.; Vukovic, V.; Oroz, K.; Coric, L.; Skoro, M.; et al. New studies with stable gastric pentadecapeptide protecting gastrointestinal tract. Significance of counteraction of vascular and multiorgan failure of occlusion/occlusion-like syndrome in cytoprotection/organoprotection. Inflammopharmacology 2024, 32, 3119–3161. [Google Scholar] [CrossRef] [PubMed]
  7. Sikiric, P.; Boban Blagaic, A.; Strbe, S.; Beketic Oreskovic, L.; Oreskovic, I.; Sikiric, S.; Staresinic, M.; Sever, M.; Kokot, A.; Jurjevic, I.; et al. The stable gastric pentadecapeptide BPC 157 pleiotropic beneficial activity and its possible relations with neurotransmitter activity. Pharmaceuticals 2024, 17, 461. [Google Scholar] [CrossRef] [PubMed]
  8. Sikiric, P.; Gojkovic, S.; Knezevic, M.; Tepes, M.; Strbe, S.; Vukojevic, J.; Duzel, A.; Kralj, T.; Krezic, I.; Zizek, H.; et al. Stable gastric pentadecapeptide BPC 157: Prompt particular activation of collateral pathways. Curr. Med. Chem. 2023, 30, 1568–1573. [Google Scholar] [CrossRef] [PubMed]
  9. Sikiric, P.; Kokot, A.; Kralj, T.; Zlatar, M.; Masnec, S.; Lazic, R.; Loncaric, K.; Oroz, K.; Sablic, M.; Boljesic, M.; et al. Stable gastric pentadecapeptide BPC 157—Possible novel therapy of glaucoma and other ocular conditions. Pharmaceuticals 2023, 16, 1052. [Google Scholar] [CrossRef] [PubMed]
  10. Staresinic, M.; Japjec, M.; Vranes, H.; Prtoric, A.; Zizek, H.; Krezic, I.; Gojkovic, S.; Smoday, I.M.; Oroz, K.; Staresinic, E.; et al. Stable gastric pentadecapeptide BPC 157 and striated, smooth, and heart muscle. Biomedicines 2022, 10, 3221. [Google Scholar] [CrossRef] [PubMed]
  11. Sikiric, P.; Udovicic, M.; Barisic, I.; Balenovic, D.; Zivanovic Posilovic, G.; Strinic, D.; Uzun, S.; Sikiric, S.; Krezic, I.; Zizek, H.; et al. Stable gastric pentadecapeptide BPC 157 as useful cytoprotective peptide therapy in the heart disturbances, myocardial infarction, heart failure, pulmonary hypertension, arrhythmias, and thrombosis presentation. Biomedicines 2022, 10, 2696. [Google Scholar] [CrossRef] [PubMed]
  12. Sikiric, P.; Skrtic, A.; Gojkovic, S.; Krezic, I.; Zizek, H.; Lovric, E.; Sikiric, S.; Knezevic, M.; Strbe, S.; Milavic, M.; et al. Cytoprotective gastric pentadecapeptide BPC 157 resolves major vessel occlusion disturbances, ischemia-reperfusion injury following Pringle maneuver, and Budd-Chiari syndrome. World J. Gastroenterol. 2022, 28, 23–46. [Google Scholar] [CrossRef] [PubMed]
  13. Robert, A. Cytoprotection by prostaglandins. Gastroenterology 1979, 77, 761–767. [Google Scholar] [CrossRef] [PubMed]
  14. Robert, A. Current history of cytoprotection. Prostaglandins 1981, 21 (Suppl. S1), 89–96. [Google Scholar] [CrossRef] [PubMed]
  15. Robert, A.; Nezamis, J.E.; Lancaster, C.; Davis, J.P.; Field, S.O.; Hanchar, A.J. Mild irritants prevent gastric necrosis through “adaptive cytoprotection” mediated by prostaglandins. Am. J. Physiol. 1983, 245, G113–G121. [Google Scholar] [CrossRef] [PubMed]
  16. Szabo, S. Role of sulfhydryls and early vascular lesions in gastric mucosal injury. Acta Physiol. Hung. 1984, 64, 203–214. [Google Scholar] [PubMed]
  17. Trier, J.S.; Szabo, S.; Allan, C.H. Ethanol-induced damage to mucosal capillaries of rat stomach. Ultrastructural features and effects of prostaglandin F2β and cysteamine. Gastroenterology 1987, 92, 13–22. [Google Scholar] [CrossRef] [PubMed]
  18. Pihan, G.; Majzoubi, D.; Haudenschild, C.; Trier, J.S.; Szabo, S. Early microcirculatory stasis in acute gastric mucosal injury in the rat and prevention by 16,16-dimethyl prostaglandin E2 or sodium thiosulfate. Gastroenterology 1986, 91, 1415–1426. [Google Scholar] [CrossRef] [PubMed]
  19. Szabo, S.; Trier, J.S.; Brown, A.; Schnoor, J. Early vascular injury and increased vascular permeability in gastric mucosal injury caused by ethanol in the rat. Gastroenterology 1985, 88 (Suppl. S1), 228–236. [Google Scholar] [CrossRef] [PubMed]
  20. Szabo, S. Experimental basis for a role for sulfhydryls and dopamine in ulcerogenesis: A primer for cytoprotection–organoprotection. Klin. Wochenschr. 1986, 64 (Suppl. S7), 116–122. [Google Scholar] [PubMed]
  21. Szabo, S.; Usadel, K.H. Cytoprotection-organoprotection by somatostatin: Gastric and hepatic lesions. Experientia 1982, 38, 254–256. [Google Scholar] [CrossRef] [PubMed]
  22. Seiwerth, S.; Milavic, M.; Vukojevic, J.; Gojkovic, S.; Krezic, I.; Vuletic, L.B.; Pavlov, K.H.; Petrovic, A.; Sikiric, S.; Vranes, H.; et al. Stable gastric pentadecapeptide BPC 157 and wound healing. Front. Pharmacol. 2021, 12, 627533. [Google Scholar] [CrossRef] [PubMed]
  23. Seiwerth, S.; Rucman, R.; Turkovic, B.; Sever, M.; Klicek, R.; Radic, B.; Drmic, D.; Stupnisek, M.; Misic, M.; Vuletic, L.B.; et al. BPC 157 and standard angiogenic growth factors. Gastrointestinal tract healing, lessons from tendon, ligament, muscle and bone healing. Curr. Pharm. Des. 2018, 24, 1972–1989. [Google Scholar] [CrossRef] [PubMed]
  24. Sikiric, P.; Drmic, D.; Sever, M.; Klicek, R.; Blagaic, A.B.; Tvrdeic, A.; Kralj, T.; Kovac, K.K.; Vukojevic, J.; Siroglavic, M.; et al. Fistulas healing. Stable gastric pentadecapeptide BPC 157 therapy. Curr. Pharm. Des. 2020, 26, 2991–3000. [Google Scholar] [CrossRef] [PubMed]
  25. Bajramagic, S.; Sever, M.; Rasic, F.; Staresinic, M.; Skrtic, A.; Beketic Oreskovic, L.; Oreskovic, I.; Strbe, S.; Loga Zec, S.; Hrabar, J.; et al. Stable gastric pentadecapeptide BPC 157 and intestinal anastomoses therapy in rats—A review. Pharmaceuticals 2024, 17, 1081. [Google Scholar] [CrossRef] [PubMed]
  26. 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, 1, CD009782. [Google Scholar] [CrossRef] [PubMed]
  27. Tripathi, R.C.; Parapuram, S.K.; Tripathi, B.J.; Zhong, Y.; Chalam, K.V. Corticosteroids and glaucoma risk. Drugs Aging 1999, 15, 439–450. [Google Scholar] [CrossRef] [PubMed]
  28. Pleyer, U.; Ursell, P.G.; Rama, P. Intraocular pressure effects of common topical steroids for post-cataract inflammation: Are they all the same? Ophthalmol. Ther. 2013, 2, 55–72. [Google Scholar] [CrossRef] [PubMed]
  29. Zarei-Ghanavati, S.; Baghdasaryan, E.; Ramirez-Miranda, A.; Nguyen, M.; Yu, F.; Lee, G.J.; Deng, S.X. Elevated intraocular pressure is a common complication during active microbial keratitis. Am. J. Ophthalmol. 2011, 152, 575–581.e1. [Google Scholar] [CrossRef] [PubMed]
  30. Sakiyalak, D.; Chattagoon, Y. Incidence of and risk factors for secondary ocular hypertension in moderate to severe infectious ulcerative keratitis. Clin. Ophthalmol. 2018, 12, 2121–2128. [Google Scholar] [CrossRef] [PubMed]
  31. Williams, D.L.; Burg, P. Comparison of intraocular pressure and tear production in healthy eyes and in eyes with corneal ulceration in dogs. Vet. Ophthalmol. 2017, 20, 206–211. [Google Scholar] [CrossRef] [PubMed]
  32. Wilson, S.E. Corneal wound healing. Exp. Eye Res. 2020, 197, 108089. [Google Scholar] [CrossRef] [PubMed]
  33. Baldwin, H.C.; Marshall, J. Growth factors in corneal wound healing following refractive surgery: A review. Acta Ophthalmol. Scand. 2002, 80, 238–247. [Google Scholar] [CrossRef] [PubMed]
  34. Nentwich, M.M.; Ulbig, M.W. Diabetic retinopathy—Ocular complications of diabetes mellitus. World J. Diabetes 2015, 6, 489–499. [Google Scholar] [CrossRef] [PubMed]
  35. Kralj, T.; Kokot, A.; Zlatar, M.; Masnec, S.; Kasnik Kovac, K.; Milkovic Perisa, M.; Batelja Vuletic, L.; Giljanovic, A.; Strbe, S.; Sikiric, S.; et al. Stable gastric pentadecapeptide BPC 157 therapy of rat glaucoma. Biomedicines 2021, 10, 89. [Google Scholar] [CrossRef] [PubMed]
  36. Kokot, A.; Zlatar, M.; Stupnisek, M.; Drmic, D.; Radic, R.; Vcev, A.; Seiwerth, S.; Sikiric, P. NO system dependence of atropine-induced mydriasis and L-NAME- and L-arginine-induced miosis: Reversal by the pentadecapeptide BPC 157 in rats and guinea pigs. Eur. J. Pharmacol. 2016, 771, 211–219. [Google Scholar] [CrossRef] [PubMed]
  37. Zlatar, M.; Kokot, A.; Vuletic, L.B.; Masnec, S.; Kralj, T.; Perisa, M.M.; Barisic, I.; Radic, B.; Milanovic, K.; Drmic, D.; et al. BPC 157 as a therapy for retinal ischemia induced by retrobulbar application of L-NAME in rats. Front. Pharmacol. 2021, 12, 632295. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Lazić, R.; Gabrić, N.; Dekaris, I.; Bosnar, D.; Boban-Blagaić, A.; Sikiric, P. Gastric pentadecapeptide BPC 157 promotes corneal epithelial defects healing in rats. Coll. Antropol. 2005, 29, 321–325. [Google Scholar] [PubMed]
  39. Kralj, T.; Kokot, A.; Kasnik, K.; Drmic, D.; Zlatar, M.; Seiwerth, S.; Sikiric, P. Effects of pentadecapeptide BPC 157 on experimental rat model of dry eye. FASEB J. 2017, 31 (Suppl. S1), 993.3. [Google Scholar] [CrossRef]
  40. Mirković, I.; Kralj, T.; Lozić, M.; Stambolija, V.; Kovačević, J.; Vrdoljak, L.; Zlatar, M.; Milanović, K.; Drmić, D.; Predović, J.; et al. Pentadecapeptide BPC 157 shortens duration of tetracaine- and oxybuprocaine-induced corneal anesthesia in rats. Acta Clin. Croat. 2020, 59, 394–406. [Google Scholar] [CrossRef] [PubMed]
  41. Stjernschantz, J.W. From PGF2α-isopropyl ester to latanoprost: A review of the development of Xalatan—The Proctor Lecture. Investig. Ophthalmol. Vis. Sci. 2001, 42, 1134–1145. [Google Scholar] [PubMed]
  42. Sikiric, P.; Buljan, M.; Vnuk, D.; Krstonijevic, Z.; Sever, M.; Lojo, N.; Drmić, D.; Zenko Sever, A.; Barić, M.; Starčević, N.; et al. Effect of pentadecapeptide BPC 157 on rotator cuff tear injury in rat. FASEB J. 2014, 28 (Suppl. S1), 844.9. [Google Scholar] [CrossRef]
  43. Krivic, A.; Anic, T.; Seiwerth, S.; Huljev, D.; Sikiric, P. Achilles detachment in rat and stable gastric pentadecapeptide BPC 157: Promoted tendon-to-bone healing and opposed corticosteroid aggravation. J. Orthop. Res. 2006, 24, 982–989. [Google Scholar] [CrossRef] [PubMed]
  44. Staresinic, M.; Sebecic, B.; Patrlj, L.; Jadrijevic, S.; Suknaic, S.; Perovic, D.; Aralica, G.; Zarkovic, N.; Borovic, S.; Srdjak, M.; et al. Gastric pentadecapeptide BPC 157 accelerates healing of transected rat Achilles tendon and in vitro stimulates tendocytes growth. J. Orthop. Res. 2003, 21, 976–983. [Google Scholar] [CrossRef] [PubMed]
  45. Krivic, A.; Majerovic, M.; Jelic, I.; Seiwerth, S.; Sikiric, P. Modulation of early functional recovery of Achilles tendon-to-bone unit after transection by BPC 157 and methylprednisolone. Inflamm. Res. 2008, 57, 205–210. [Google Scholar] [CrossRef] [PubMed]
  46. Japjec, M.; Horvat Pavlov, K.; Petrovic, A.; Staresinic, M.; Sebecic, B.; Buljan, M.; Vranes, H.; Giljanovic, A.; Drmic, D.; Japjec, M.; et al. Stable gastric pentadecapeptide BPC 157 as a therapy for the disable myotendinous junctions in rats. Biomedicines 2021, 9, 1547. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Matek, D.; Matek, I.; Staresinic, E.; Japjec, M.; Bojanic, I.; Boban Blagaic, A.; Beketic Oreskovic, L.; Oreskovic, I.; Ziger, T.; Novinscak, T.; et al. Stable gastric pentadecapeptide BPC 157 as therapy after surgical detachment of the quadriceps muscle from its attachments for muscle-to-bone reattachment in rats. Pharmaceutics 2025, 17, 119. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  48. Cerovecki, T.; Bojanic, I.; Brcic, L.; Radic, B.; Vukoja, I.; Seiwerth, S.; Sikiric, P. Pentadecapeptide BPC 157 (PL 14736) improves ligament healing in the rat. J. Orthop. Res. 2010, 28, 1155–1161. [Google Scholar] [CrossRef] [PubMed]
  49. Brcic, L.; Brcic, I.; Staresinic, M.; Novinscak, T.; Sikiric, P.; Seiwerth, S. Modulatory effect of gastric pentadecapeptide BPC 157 on angiogenesis in muscle and tendon healing. J. Physiol. Pharmacol. 2009, 60 (Suppl. S7), 191–196. [Google Scholar] [PubMed]
  50. Feizi, S. Corneal endothelial cell dysfunction: Etiologies and management. Ther. Adv. Ophthalmol. 2018, 10, 2515841418815802. [Google Scholar] [CrossRef] [PubMed]
  51. Aloe, L.; Tirassa, P.; Lambiase, A. The topical application of nerve growth factor as a pharmacological tool for human corneal and skin ulcers. Pharmacol. Res. 2008, 57, 253–258. [Google Scholar] [CrossRef] [PubMed]
  52. Lambiase, A.; Manni, L.; Bonini, S.; Rama, P.; Micera, A.; Aloe, L. Nerve growth factor promotes corneal healing: Structural, biochemical, and molecular analyses of rat and human corneas. Investig. Ophthalmol. Vis. Sci. 2000, 41, 1063–1069. [Google Scholar] [PubMed]
  53. Bentley, E.; Murphy, C.J. Topical therapeutic agents that modulate corneal wound healing. Vet. Clin. N. Am. Small Anim. Pract. 2004, 34, 623–638. [Google Scholar] [CrossRef]
  54. Gong, J.; Ding, G.; Hao, Z.; Li, Y.; Deng, A.; Zhang, C. Elucidating the mechanism of corneal epithelial cell repair: Unraveling the impact of growth factors. Front. Med. 2024, 11, 1384500. [Google Scholar] [CrossRef]
  55. Schultz, G.S.; Chegini, N.; Grant, M.; Khaw, P.T.; MacKay, S.L.D. Effect of growth factors on corneal wound healing. Acta Ophthalmol. 1992, 70, 60–66. [Google Scholar] [CrossRef]
  56. Drzyzga, Ł.; Spiewak, D.; Dorecka, M.; Wygledowska-Promienska, D. Available therapeutic options for corneal neovascularization: A review. Int. J. Mol. Sci. 2024, 25, 5479. [Google Scholar] [CrossRef]
  57. Maddula, S.; Davis, D.K.; Maddula, S.; Burrow, M.K.; Ambati, B.K. Horizons in therapy for corneal angiogenesis. Ophthalmology 2011, 118, 591–599. [Google Scholar] [CrossRef] [PubMed]
  58. Ellenberg, D.; Azar, D.T.; Hallak, J.A.; Tobaigy, F.; Han, K.Y.; Jain, S.; Zhou, Z.; Chang, J.-H. Novel aspects of corneal angiogenic and lymphangiogenic privilege. Prog. Retin. Eye Res. 2010, 29, 208–248. [Google Scholar] [CrossRef]
  59. Nicholas, M.P.; Mysore, N. Corneal neovascularization. Exp. Eye Res. 2021, 202, 108363. [Google Scholar] [CrossRef]
  60. Ung, L.; Chodosh, J. Urgent unmet needs in the care of bacterial keratitis: An evidence-based synthesis. Ocul. Surf. 2023, 28, 378–400. [Google Scholar] [CrossRef] [PubMed]
  61. Heinz, C.; Bograd, N.; Koch, J.; Heiligenhaus, A. Ocular hypertension and glaucoma incidence in patients with scleritis. Graefes Arch. Clin. Exp. Ophthalmol. 2013, 251, 139–142. [Google Scholar] [CrossRef] [PubMed]
  62. Ruenzi, M.; Stolte, M.; Veljaca, M.; Oreskovic, K.; Peterson, J.; Ulcerative Colitis Study Group. A multicenter, randomized, double blind, placebo-controlled phase II study of PL 14736 enema in the treatment of mild-to-moderate ulcerative colitis. Gastroenterology 2005, 128, A584. [Google Scholar] [CrossRef]
  63. Veljaca, M.; Pavic-Sladoljev, D.; Mildner, B.; Brajsa, K.; Bubenik, M.; Stipanicic, S.; Parnham, M. Safety, tolerability and pharmacokinetics of PL14736, a novel agent for treatment of ulcerative colitis, in healthy male volunteers. Gut 2003, 51 (Suppl. SIII), A309. [Google Scholar] [CrossRef]
  64. Lee, E.; Padgett, B. Intra-articular injection of BPC 157 for multiple types of knee pain. Altern. Ther. Health Med. 2021, 27, 8–13. [Google Scholar]
  65. Lee, E.; Walker, C.; Ayadi, B. Effect of BPC-157 on symptoms in patients with interstitial cystitis: A pilot study. Altern. Ther. Health Med. 2024, 30, 12–17. [Google Scholar] [PubMed]
  66. Vasireddi, N.; Hahamyan, H.; Salata, M.J.; Karns, M.; Calcei, J.G.; Voos, J.E.; Apostolakos, J.M. Emerging use of BPC-157 in orthopaedic sports medicine: A systematic review. HSS J. 2025; in press. [Google Scholar] [CrossRef] [PubMed]
  67. McGuire, F.P.; Martinez, R.; Lenz, A.; Skinner, L.; Cushman, D.M. Regeneration or risk? A narrative review of BPC-157 for musculoskeletal healing. Curr. Rev. Musculoskelet. Med. 2025; in press. [Google Scholar] [CrossRef] [PubMed]
  68. Veljaca, M.; Chan, K.; Guglietta, A. Digestion of h-EGF, h-TGF alpha, and BPC-15 in human gastric juice. Gastroenterology 1995, 108, 761. [Google Scholar] [CrossRef]
  69. Tkalcević, V.I.; Cuzić, S.; Brajsa, K.; Mildner, B.; Bokulić, A.; Situm, K.; Perović, D.; Glojnarić, I.; Parnham, M.J. Enhancement by PL 14736 of granulation and collagen organization in healing wounds and the potential role of egr-1 expression. Eur. J. Pharmacol. 2007, 570, 212–221. [Google Scholar] [CrossRef]
  70. Wu, H.; Wei, M.; Li, N.; Lu, Q.; Shrestha, S.M.; Tan, J.; Zhang, Z.; Wu, G.; Shi, R. Clopidogrel-induced gastric injury in rats is attenuated by stable gastric pentadecapeptide BPC 157. Drug Des. Devel. Ther. 2020, 14, 5599–5610. [Google Scholar] [CrossRef]
  71. Hsieh, M.J.; Lee, C.H.; Chueh, H.Y.; Chang, G.J.; Huang, H.Y.; Lin, Y.; Pang, J.S. Modulatory effects of BPC 157 on vasomotor tone and the activation of Src-Caveolin-1-endothelial nitric oxide synthase pathway. Sci. Rep. 2020, 10, 17078. [Google Scholar] [CrossRef]
  72. Hsieh, M.J.; Liu, H.T.; Wang, C.N.; Huang, H.Y.; Lin, Y.; Ko, Y.S.; Wang, J.S.; Chang, V.H.; Pang, J.S. Therapeutic potential of pro-angiogenic BPC 157 is associated with VEGFR2 activation and up-regulation. J. Mol. Med. 2017, 95, 323–333. [Google Scholar] [CrossRef]
  73. Radeljak, S.; Seiwerth, S.; Sikiric, P. BPC 157 inhibits cell growth and VEGF signalling via the MAPK kinase pathway in the human melanoma cell line. Melanoma Res. 2004, 14, A14–A15. [Google Scholar] [CrossRef]
  74. Huang, B.S.; Huang, S.C.; Chen, F.H.; Chang, Y.; Mei, H.F.; Huang, H.Y.; Chen, W.Y.; Pang, J.S. Pentadecapeptide BPC 157 efficiently reduces radiation-induced liver injury and lipid accumulation through Kruppel-like factor 4 upregulation both in vivo and in vitro. Life Sci. 2022, 310, 121072. [Google Scholar] [CrossRef]
  75. Wang, X.Y.; Qu, M.; Duan, R.; Shi, D.; Jin, L.; Gao, J.; Wood, J.D.; Li, J.; Wang, G.D. Cytoprotective mechanism of the novel gastric peptide in gastrointestinal tract and cultured enteric neurons and glial cells. Neurosci. Bull. 2019, 35, 167–170. [Google Scholar] [CrossRef]
  76. Chang, C.H.; Tsai, W.C.; Hsu, Y.H.; Pang, J.H.S. Pentadecapeptide BPC 157 enhances the growth hormone receptor expression in tendon fibroblasts. Molecules 2014, 19, 19066–19077. [Google Scholar] [CrossRef] [PubMed]
  77. Chang, C.H.; Tsai, W.C.; Lin, M.S.; Hsu, Y.H.; Pang, J.H.S. The promoting effect of pentadecapeptide BPC 157 on tendon healing involves tendon outgrowth, cell survival, and cell migration. J. Appl. Physiol. 2011, 110, 774–780. [Google Scholar] [CrossRef] [PubMed]
  78. Park, J.M.; Lee, H.J.; Sikiric, P.; Hahm, K.B. BPC 157 rescued NSAID-cytotoxicity via stabilizing intestinal permeability and enhancing cytoprotection. Curr. Pharm. Des. 2020, 26, 2971–2981. [Google Scholar] [CrossRef]
  79. Kang, E.A.; Han, Y.M.; An, J.M.; Park, Y.J.; Sikiric, P.; Kim, D.H.; Kwon, K.A.; Kim, Y.J.; Yang, D.; Tchah, H.; et al. BPC 157 as potential agent rescuing from cancer cachexia. Curr. Pharm. Des. 2018, 24, 1947–1956. [Google Scholar] [CrossRef]
  80. Huang, T.; Zhang, K.; Sun, L.; Xue, X.; Zhang, C.; Shu, Z.; Mu, N.; Gu, J.; Zhang, W.; Wang, Y.; et al. Body protective compound-157 enhances alkali-burn wound healing in vivo and promotes proliferation, migration, and angiogenesis in vitro. Drug Des. Devel. Ther. 2015, 9, 2485–2499. [Google Scholar] [CrossRef] [PubMed]
  81. Sikirić, P.; Seiwerth, S.; Grabarević, Z.; Rucman, R.; Petek, M.; Jagić, V.; Turković, B.; Rotkvić, I.; Mise, S.; Zoricić, I.; et al. The influence of a novel pentadecapeptide, BPC 157, on N(G)-nitro-L-arginine methylester and L-arginine effects on stomach mucosa integrity and blood pressure. Eur. J. Pharmacol. 1997, 332, 23–33. [Google Scholar] [CrossRef] [PubMed]
  82. Turkovic, B.; Sikiric, P.; Seiwerth, S.; Mise, S.; Anic, T.; Petek, M. Stable gastric pentadecapeptide BPC 157 studied for inflammatory bowel disease (PLD-116, PL14736, Pliva) induces nitric oxide synthesis. Gastroenterology 2004, 126, 287. [Google Scholar] [CrossRef]
  83. Stupnisek, M.; Kokot, A.; Drmic, D.; Hrelec-Patrlj, M.; Zenko-Sever, A.; Kolenc, D.; Radic, B.; Suran, J.; Bojic, D.; Vcev, A.; et al. Pentadecapeptide BPC 157 reduces bleeding and thrombocytopenia after amputation in rats treated with heparin, warfarin, L-NAME and L-arginine. PLoS ONE 2015, 10, e0123454. [Google Scholar] [CrossRef] [PubMed]
  84. Sikiric, P.; Drmic, D.; Boban-Blagaic, A.; Tvrdeic, A.; Krezic, I.; Gojkovic, S.; Zizek, H.; Sikiric, S.; Strbe, S.; Smoday, I.M.; et al. Stable gastric pentadecapeptide BPC 157 and NO-system. In Nitric Oxide: From Research to Therapeutics, Advances in Biochemistry in Health and Disease; Ray, A., Gulati, K., Eds.; Springer Nature: Cham, Switzerland, 2023; pp. 349–375. [Google Scholar]
  85. Sikiric, P.; Seiwerth, S.; Rucman, R.; Turkovic, B.; Rokotov, D.S.; Brcic, L.; Sever, M.; Klicek, R.; Radic, B.; Drmic, D.; et al. Stable gastric pentadecapeptide BPC 157-NO-system relation. Curr. Pharm. Des. 2014, 20, 1126–1135. [Google Scholar] [CrossRef]
  86. Mohan, M.; Natarajan, R.; Kaur, K.; Gurnani, B. Treatment approach to corneal ulcer. TNOA J. Ophthalmic Sci. Res. 2023, 61, 396–407. [Google Scholar] [CrossRef]
  87. Pfister, R.R.; Paterson, C.A.; Hayes, S.A. Topical ascorbate decreases the incidence of corneal ulceration after experimental alkali burns. Investig. Ophthalmol. Vis. Sci. 1978, 17, 1019–1024. [Google Scholar]
  88. Pfister, R.R.; Paterson, C.A. Ascorbic acid in the treatment of alkali burns of the eye. Ophthalmology 1980, 87, 1050–1057. [Google Scholar] [CrossRef]
  89. Pfister, R.R.; Paterson, C.A.; Hayes, S.A. Effects of topical 10% ascorbate solution on established corneal ulcers after severe alkali burns. Investig. Ophthalmol. Vis. Sci. 1982, 22, 382–385. [Google Scholar]
  90. Petroutsos, G.; Pouliquen, Y. Effect of ascorbic acid on ulceration in alkali-burned corneas. Ophthalmic Res. 1984, 16, 185–189. [Google Scholar] [CrossRef] [PubMed]
  91. Kasetsuwan, N.; Wu, F.M.; Hsieh, F.; Sanchez, D.; McDonnell, P.J. Effect of topical ascorbic acid on free radical tissue damage and inflammatory cell influx in the cornea after excimer laser corneal surgery. Arch. Ophthalmol. 1999, 117, 649–652. [Google Scholar] [CrossRef]
  92. Phan, T.M.M.; Zelt, R.P.; Kenyon, K.R.; Chakrabarti, B.; Foster, C.S. Ascorbic acid therapy in a thermal burn model of corneal ulceration in rabbits. Am. J. Ophthalmol. 1985, 99, 74–82. [Google Scholar] [CrossRef]
  93. Chen, J.; Lan, J.; Liu, D.; Backman, L.J.; Zhang, W.; Zhou, Q.; Danielson, P. Ascorbic acid promotes the stemness of corneal epithelial stem/progenitor cells and accelerates epithelial wound healing in the cornea. Stem Cells Transl. Med. 2017, 6, 1356–1365. [Google Scholar] [CrossRef]
  94. Li, M.; Chen, Z.; Liu, L.; Ma, X.; Zou, J. Topical vitamin C promotes the recovery of corneal alkali burns in mice. J. Ophthalmol. 2021, 2021, 2406646. [Google Scholar] [CrossRef]
  95. Hsueh, Y.-J.; Meir, Y.-J.J.; Yeh, L.-K.; Chen, W.-L.; Chen, S.-Y.; Wang, I.-J.; Hu, F.-R.; Chen, P.-L. Topical ascorbic acid ameliorates oxidative stress-induced corneal endothelial damage via suppression of apoptosis and autophagic flux blockage. Cells 2020, 9, 943. [Google Scholar] [CrossRef] [PubMed]
  96. Csorba, A.; Katona, G.; Budai-Szűcs, M.; Kovács, K.; Pál, S.; Fenyvesi, F.; Váradi, J.; Szente, L.; Fehér, P.; Vecsernyés, M.; et al. Effect of liposomal formulation of ascorbic acid on corneal permeability. Sci. Rep. 2023, 13, 3448. [Google Scholar] [CrossRef] [PubMed]
  97. Omeroğlu, S.; Peker, T.; Türközkan, N.; Omeroğlu, H. High-dose vitamin C supplementation accelerates the Achilles tendon healing in healthy rats. Arch. Orthop. Trauma Surg. 2009, 129, 281–286. [Google Scholar] [CrossRef]
  98. Noriega-González, D.C.; Drobnic, F.; Caballero-García, A.; Roche, E.; Perez-Valdecantos, D.; Córdova, A. Effect of vitamin C on tendinopathy recovery: A scoping review. Nutrients 2022, 14, 2663. [Google Scholar] [CrossRef] [PubMed]
  99. Oliva, F.; Maffulli, N.; Gissi, C.; Veronesi, F.; Calciano, L.; Fini, M.; Brogini, S.; Gallorini, M.; Passeri, C.A.L.; Bernardini, R.; et al. Combined ascorbic acid and T3 produce better healing compared to bone marrow mesenchymal stem cells in an Achilles tendon injury rat model: A proof of concept study. J. Orthop. Surg. Res. 2019, 14, 54. [Google Scholar] [CrossRef]
  100. Evrova, O.; Kellenberger, D.; Calcagni, M.; Vogel, V.; Buschmann, J. Supporting cell-based tendon therapy: Effect of PDGF-BB and ascorbic acid on rabbit Achilles tenocytes in vitro. Int. J. Mol. Sci. 2020, 21, 458. [Google Scholar] [CrossRef]
  101. Çelik, M.; Bayrak, A.; Duramaz, A.; Başaran, S.H.; Kızılkaya, C.; Kural, C.; Büyükkurt, C.D.; Erçin, E.; Erdil, M.; Aydın, B.K.; et al. The effect of fibrin clot and C vitamin on the surgical treatment of Achilles tendon injury in the rat model. Foot Ankle Surg. 2021, 27, 681–687. [Google Scholar] [CrossRef]
  102. Lui, P.P.Y.; Wong, O.T.; Lee, Y.W.; Tsui, A.Y.; Yung, P.S.; Chan, K.M. Transplantation of tendon-derived stem cells pre-treated with connective tissue growth factor and ascorbic acid in vitro promoted better tendon repair in a patellar tendon window injury rat model. Cytotherapy 2016, 18, 99–112. [Google Scholar] [CrossRef]
  103. Hung, L.K.; Fu, S.C.; Lee, Y.W.; Mok, T.Y.; Chan, K.M. Local vitamin C injection reduced tendon adhesion in a chicken model of flexor digitorum profundus tendon injury. J. Bone Jt. Surg. Am. 2013, 95, e41. [Google Scholar] [CrossRef]
  104. Phan, T.M.; Foster, C.S.; Shaw, C.D.; Zagachin, L.M.; Colvin, R.B. Topical fibronectin in an alkali burn model of corneal ulceration in rabbits. Arch. Ophthalmol. 1991, 109, 414–419. [Google Scholar] [CrossRef] [PubMed]
  105. Nishida, T. The role of fibronectin in corneal wound healing explored by a physician-scientist. Jpn. J. Ophthalmol. 2012, 56, 417–431. [Google Scholar] [CrossRef] [PubMed]
  106. Nakamura, M.; Nishida, T. Synergistic effects of hyaluronan and fibronectin on epithelial migration in rabbit cornea in vitro. Cornea 1999, 18, 686–692. [Google Scholar] [CrossRef] [PubMed]
  107. Nakamura, M.; Sato, N.; Chikama, T.; Hasegawa, Y.; Nishida, T. Fibronectin facilitates corneal epithelial wound healing in diabetic rats. Exp. Eye Res. 1997, 64, 355–359. [Google Scholar] [CrossRef] [PubMed]
  108. Phan, T.M.; Foster, C.S.; Zagachin, L.M.; Colvin, R.B. Role of fibronectin in the healing of superficial keratectomies in vitro. Investig. Ophthalmol. Vis. Sci. 1989, 30, 386–391. [Google Scholar] [PubMed]
  109. Phan, T.M.; Foster, C.S.; Wasson, P.J.; Fujikawa, L.S.; Zagachin, L.M.; Colvin, R.B. Role of fibronectin and fibrinogen in healing of corneal epithelial scrape wounds. Investig. Ophthalmol. Vis. Sci. 1989, 30, 377–385. [Google Scholar] [PubMed]
  110. Kim, K.S.; Oh, J.S.; Kim, I.S.; Jo, J.S. Topical fibronectin treatment in persistent corneal epithelial defects and corneal ulcers. Korean J. Ophthalmol. 1990, 4, 5–11. [Google Scholar] [CrossRef] [PubMed]
  111. Nishida, T.; Nakagawa, S.; Nishibayashi, C.; Tanaka, H.; Manabe, R. Fibronectin enhancement of corneal epithelial wound healing of rabbits in vivo. Arch. Ophthalmol. 1984, 102, 455–456. [Google Scholar] [CrossRef] [PubMed]
  112. Phan, T.M.; Foster, C.S.; Boruchoff, S.A.; Zagachin, L.M.; Colvin, R.B. Topical fibronectin in the treatment of persistent corneal epithelial defects and trophic ulcers. Am. J. Ophthalmol. 1987, 104, 494–501. [Google Scholar] [CrossRef] [PubMed]
  113. Frangieh, G.T.; Hayashi, K.; Teekhasaenee, C.; Wolf, G.; Colvin, R.B.; Gipson, I.K.; Kenyon, K.R. Fibronectin and corneal epithelial wound healing in the vitamin A-deficient rat. Arch. Ophthalmol. 1989, 107, 567–571. [Google Scholar] [CrossRef] [PubMed]
  114. Watanabe, K.; Frangieh, G.; Reddy, C.V.; Kenyon, K.R. Effect of fibronectin on corneal epithelial wound healing in the vitamin A-deficient rat. Investig. Ophthalmol. Vis. Sci. 1991, 32, 2159–2162. [Google Scholar] [PubMed]
  115. Spigelman, A.V.; Deutsch, T.A.; Sugar, J. Application of homologous fibronectin to persistent human corneal epithelial defects. Cornea 1987, 6, 128–130. [Google Scholar] [CrossRef] [PubMed]
  116. McCulley, J.P.; Horowitz, B.; Husseini, Z.M.; Horowitz, M. Fibronectin Study Group. Topical fibronectin therapy of persistent corneal epithelial defects. Trans. Am. Ophthalmol. Soc. 1993, 91, 367–386. [Google Scholar] [PubMed] [PubMed Central]
  117. Gordon, J.F.; Johnson, P.; Musch, D.C. Chiron Vision Fibronectin Study Group. Topical fibronectin ophthalmic solution in the treatment of persistent defects of the corneal epithelium. Am. J. Ophthalmol. 1995, 119, 281–287. [Google Scholar] [CrossRef] [PubMed]
  118. Muether, P.S.; Dell, S.; Kociok, N.; Zahn, G.; Stragies, R.; Vossmeyer, D.; Joussen, A.M. The role of integrin α5β1 in the regulation of corneal neovascularization. Exp. Eye Res. 2007, 85, 356–365. [Google Scholar] [CrossRef] [PubMed]
  119. Birchler, M.; Viti, F.; Zardi, L.; Spiess, B.; Neri, D. Selective targeting and photocoagulation of ocular angiogenesis mediated by a phage-derived human antibody fragment. Nat. Biotechnol. 1999, 17, 984–988. [Google Scholar] [CrossRef] [PubMed]
  120. Grant, M.B.; Caballero, S.; Bush, D.M.; Spoerri, P.E. Fibronectin fragments modulate human retinal capillary cell proliferation and migration. Diabetes 1998, 47, 1335–1340. [Google Scholar] [CrossRef] [PubMed]
  121. Barbariga, M.; Vallone, F.; Mosca, E.; Bignami, F.; Magagnotti, C.; Fonteyne, P.; Chiappori, F.; Milanesi, L.; Rama, P.; Andolfo, A.; et al. The role of extracellular matrix in mouse and human corneal neovascularization. Sci. Rep. 2019, 9, 14272. [Google Scholar] [CrossRef]
  122. Faralli, J.A.; Filla, M.S.; Peters, D.M. Role of fibronectin in primary open angle glaucoma. Cells 2019, 8, 1518. [Google Scholar] [CrossRef] [PubMed]
  123. Roberts, A.L.; Mavlyutov, T.; Perlmutter, T.E.; Curry, S.M.; Harris, S.L.; Chauhan, A.K.; McDowell, C.M. Fibronectin extra domain A (FN-EDA) elevates intraocular pressure through Toll-like receptor 4 signaling. Sci. Rep. 2020, 10, 9815. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  124. Faralli, J.A.; Filla, M.S.; McDowell, C.M.; Peters, D.M. Disruption of fibronectin fibrillogenesis affects intraocular pressure (IOP) in BALB/cJ mice. PLoS ONE 2020, 15, e0237932. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  125. Mavlyutov, T.A.; Myrah, J.J.; Chauhan, A.K.; Liu, Y.; McDowell, C.M. Fibronectin extra domain A (FN-EDA) causes glaucomatous trabecular meshwork, retina, and optic nerve damage in mice. Cell Biosci. 2022, 12, 72. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  126. Medina-Ortiz, W.E.; Belmares, R.; Neubauer, S.; Wordinger, R.J.; Clark, A.F. Cellular fibronectin expression in human trabecular meshwork and induction by transforming growth factor-β2. Investig. Ophthalmol. Vis. Sci. 2013, 54, 6779–6788. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  127. Dismuke, W.M.; Klingeborn, M.; Stamer, W.D. Mechanism of fibronectin binding to human trabecular meshwork exosomes and its modulation by dexamethasone. PLoS ONE 2016, 11, e0165326. [Google Scholar] [CrossRef] [PubMed]
  128. Kasetti, R.B.; Maddineni, P.; Millar, J.C.; Clark, A.F.; Zode, G.S. Increased synthesis and deposition of extracellular matrix proteins leads to endoplasmic reticulum stress in the trabecular meshwork. Sci. Rep. 2017, 7, 14951. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  129. Munuera, I.; Aragon-Navas, A.; Villacampa, P.; Gonzalez-Cela, M.A.; Subías, M.; Pablo, L.E.; Garcia-Feijoo, J.; Herrero-Vanrell, R.; Garcia-Martin, E.; Bravo-Osuna, I.; et al. Chronic glaucoma induced in rats by a single injection of fibronectin-loaded PLGA microspheres: IOP-dependent and IOP-independent neurodegeneration. Int. J. Mol. Sci. 2023, 25, 9. [Google Scholar] [CrossRef] [PubMed]
  130. Docheva, D.; Müller, S.A.; Majewski, M.; Evans, C.H. Biologics for tendon repair. Adv. Drug Deliv. Rev. 2015, 84, 222–239. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  131. Fraser, J.R.E.; Laurent, T.C.; Laurent, U.B. Hyaluronan: Its nature, distribution, functions and turnover. J. Intern. Med. 1997, 242, 27–33. [Google Scholar] [CrossRef] [PubMed]
  132. Shaharudin, A.; Aziz, Z. Effectiveness of hyaluronic acid and its derivatives on chronic wounds: A systematic review. J. Wound Care 2016, 25, 585–592. [Google Scholar] [CrossRef] [PubMed]
  133. Carlson, E.C.; Ousler, G.W., 3rd; Hepworth, L.A.; Semba, C.P. Impact of hyaluronic acid–containing artificial tear products on reepithelialization in an in vivo corneal wound model. Cornea 2018, 37, 893–897. [Google Scholar] [CrossRef] [PubMed]
  134. Chung, J.-H.; Park, Y.-K.; Paek, S.-M.; Chong, Y.-H.; Kim, W.-K. Effect of Na-hyaluronan on stromal and endothelial healing in experimental corneal alkali wounds. Ophthalmic Res. 1999, 31, 432–439. [Google Scholar] [CrossRef] [PubMed]
  135. Chang, W.H.; Liu, P.Y.; Lin, M.H.; Lu, C.J.; Chou, H.Y.; Nian, C.Y.; Jiang, Y.T.; Hsu, Y.H. Applications of hyaluronic acid in ophthalmology and contact lenses. Molecules 2021, 26, 2485. [Google Scholar] [CrossRef] [PubMed]
  136. Lin, T.; Gong, L. Sodium hyaluronate eye drops treatment for superficial corneal abrasion caused by mechanical damage: A randomized clinical trial in the People’s Republic of China. Drug Des. Dev. Ther. 2015, 9, 687–694. [Google Scholar] [CrossRef] [PubMed]
  137. Ho, W.T.; Chiang, T.H.; Chang, S.W.; Chen, Y.H.; Hu, F.R.; Wang, I.J. Enhanced corneal wound healing with hyaluronic acid and high-potassium artificial tears. Clin. Exp. Optom. 2013, 96, 536–541. [Google Scholar] [CrossRef] [PubMed]
  138. Nakamura, M.; Sato, N.; Chikama, T.I.; Hasegawa, Y.; Nishida, T. Hyaluronan facilitates corneal epithelial wound healing in diabetic rats. Exp. Eye Res. 1997, 64, 1043–1050. [Google Scholar] [CrossRef] [PubMed]
  139. Turley, E.A.; Noble, P.W.; Bourguignon, L.Y. Signaling properties of hyaluronan receptors. J. Biol. Chem. 2002, 277, 4589–4592. [Google Scholar] [CrossRef] [PubMed]
  140. Yang, G.; Espandar, L.; Mamalis, N.; Prestwich, G.D. A cross-linked hyaluronan gel accelerates healing of corneal epithelial abrasion and alkali burn injuries in rabbits. Vet. Ophthalmol. 2010, 13, 144–150. [Google Scholar] [CrossRef] [PubMed]
  141. Camillieri, G.; Bucolo, C.; Rossi, S.; Drago, F. Hyaluronan-induced stimulation of corneal wound healing is a pure pharmacological effect. J. Ocul. Pharmacol. Ther. 2004, 20, 548–553. [Google Scholar] [CrossRef] [PubMed]
  142. Chaidaroon, W.; Satayawut, N.; Tananuvat, N. Effect of 2% hyaluronic acid on the rate of healing of corneal epithelial defect after pterygium surgery: A randomized controlled trial. Drug Des. Dev. Ther. 2021, 15, 4435–4443. [Google Scholar] [CrossRef] [PubMed]
  143. Ciccone, V.; Zazzetta, M.; Morbidelli, L. Comparison of the effect of two hyaluronic acid preparations on fibroblast and endothelial cell functions related to angiogenesis. Cells 2019, 8, 1479. [Google Scholar] [CrossRef] [PubMed]
  144. Park, S.K.; Ha, M.; Kim, E.J.; Seo, Y.A.; Lee, H.J.; Myung, D.; Kim, H.S.; Na, K.S. Hyaluronic acid hydrogels crosslinked via blue light-induced thiol-ene reaction for the treatment of rat corneal alkali burn. Regen. Ther. 2022, 20, 51–60. [Google Scholar] [CrossRef] [PubMed]
  145. Kaux, J.F.; Samson, A.; Crielaard, J.M. Hyaluronic acid and tendon lesions. Muscles Ligaments Tendons J. 2016, 5, 264–269. [Google Scholar] [CrossRef] [PubMed]
  146. Gaughan, E.M.; Nixon, A.J.; Krook, L.P.; Yeager, A.E.; Mann, K.A.; Mohammed, H.; Bartel, D.J. Effects of sodium hyaluronate on tendon healing and adhesion formation in horses. Am. J. Vet. Res. 1991, 52, 764–773. [Google Scholar] [CrossRef] [PubMed]
  147. Dincel, Y.M.; Adanir, O.; Arikan, Y.; Caglar, A.K.; Dogru, S.C.; Arslan, Y.Z. Effects of high-dose vitamin C and hyaluronic acid on tendon healing. Acta Ortop. Bras. 2018, 26, 82–85. [Google Scholar] [CrossRef] [PubMed]
  148. Hsu, C.-H.; Chen, P.; Yang, C.-P.; Chuang, C.-A.; Chan, Y.-S.; Chiu, J.C.-H. The results of preventing postoperative Achilles tendon adhesion using cross-linked and non-cross-linked hyaluronic acid, a study with rat model. J. Orthop. Surg. Res. 2024, 19, 457. [Google Scholar] [CrossRef] [PubMed]
  149. Kang, H.J.; Subramanian, S.A.; Song, S.Y.; Hwang, J.; Lee, C.; Kim, S.J. Epigallocatechin-3-gallate (EGCG)-loaded hyaluronic acid hydrogel seems to be effective in a rat model of collagenase-induced Achilles tendinopathy. J. Funct. Biomater. 2025, 16, 55. [Google Scholar] [CrossRef] [PubMed]
  150. Benozzi, J.; Nahum, L.P.; Campanelli, J.L.; Rosenstein, R.E. Effect of hyaluronic acid on intraocular pressure in rats. Investig. Ophthalmol. Vis. Sci. 2002, 43, 2196–2200. [Google Scholar] [PubMed]
  151. Noailles, A.; Kutsyr, O.; Mayordomo-Febrer, A.; Lax, P.; López-Murcia, M.; Sanz-González, S.M.; Pinazo-Durán, M.D.; Cuenca, N. Sodium hyaluronate-induced ocular hypertension in rats damages the direction-selective circuit and inner/outer retinal plexiform layers. Investig. Ophthalmol. Vis. Sci. 2022, 63, 2. [Google Scholar] [CrossRef] [PubMed]
  152. Rainer, G.; Menapace, R.; Schmid, K.E.; Sacu, S.; Kiss, B.; Heinze, G.; Findl, O. Natural course of intraocular pressure after cataract surgery with sodium chondroitin sulfate 4%-sodium hyaluronate 3% (Viscoat). Ophthalmology 2005, 112, 1714–1718. [Google Scholar] [CrossRef] [PubMed]
  153. García-López, C.; Rodríguez-Calvo-de-Mora, M.; Borroni, D.; Sánchez-González, J.M.; Romano, V.; Rocha-de-Lossada, C. The role of matrix metalloproteinases in infectious corneal ulcers. Surv. Ophthalmol. 2023, 68, 929–939. [Google Scholar] [CrossRef]
  154. Nishida, T.; Sugioka, K.; Fukuda, K.; Murakami, J. Pivotal role of corneal fibroblasts in progression to corneal ulcer in bacterial keratitis. Int. J. Mol. Sci. 2021, 22, 8979. [Google Scholar] [CrossRef] [PubMed]
  155. Austin, A.; Lietman, T.; Rose-Nussbaumer, J. Update on the management of infectious keratitis. Ophthalmology 2017, 124, 1678–1689. [Google Scholar] [CrossRef] [PubMed]
  156. Kato, T.; Saika, S.; Ohnishi, Y. Effects of the matrix metalloproteinase inhibitor GM6001 on the destruction and alteration of epithelial basement membrane during the healing of post-alkali burn in rabbit cornea. Jpn. J. Ophthalmol. 2006, 50, 90–95. [Google Scholar] [CrossRef] [PubMed]
  157. Saika, S.; Miyamoto, T.; Yamanaka, O.; Kato, T.; Ohnishi, Y.; Flanders, K.C.; Ikeda, K.; Nakajima, Y.; Kao, W.W.; Sato, M.; et al. Therapeutic effect of topical administration of SN50, an inhibitor of nuclear factor-kappaB, in treatment of corneal alkali burns in mice. Am. J. Pathol. 2005, 166, 1393–1403. [Google Scholar] [CrossRef]
  158. Wentworth, J.S.; Paterson, C.A.; Gray, R.D. Effect of a metalloproteinase inhibitor on established corneal ulcers after an alkali burn. Investig. Ophthalmol. Vis. Sci. 1992, 33, 2174–2179. [Google Scholar]
  159. Brooks, D.E.; Ollivier, F.J. Matrix metalloproteinase inhibition in corneal ulceration. Vet. Clin. N. Am. Small Anim. Pract. 2004, 34, 611–622. [Google Scholar] [CrossRef]
  160. Wolf, M.; Clay, S.M.; Zheng, S.; Pan, P.; Chan, M.F. MMP12 inhibits corneal neovascularization and inflammation through regulation of CCL2. Sci. Rep. 2019, 9, 11579. [Google Scholar] [CrossRef]
  161. Del Buono, A.; Oliva, F.; Osti, L.; Maffulli, N. Metalloproteases and tendinopathy. Muscles Ligaments Tendons J. 2013, 3, 51–57. [Google Scholar] [CrossRef]
  162. Weinreb, R.N.; Robinson, M.R.; Dibas, M.; Stamer, W.D. Matrix metalloproteinases and glaucoma treatment. J. Ocul. Pharmacol. Ther. 2020, 36, 208–228. [Google Scholar] [CrossRef]
  163. Atanasovska Velkovska, M.; Goričar, K.; Blagus, T.; Dolžan, V.; Cvenkel, B. Association of matrix metalloproteinases polymorphisms with glaucoma risk, glaucoma phenotype, and response to treatment with selective laser trabeculoplasty or latanoprost. Int. J. Mol. Sci. 2024, 25, 13464. [Google Scholar] [CrossRef]
  164. De Groef, L.; Van Hove, I.; Dekeyster, E.; Stalmans, I.; Moons, L. MMPs in the neuroretina and optic nerve: Modulators of glaucoma pathogenesis and repair? Investig. Ophthalmol. Vis. Sci. 2014, 55, 1953–1964. [Google Scholar] [CrossRef]
  165. De Groef, L.; Van Hove, I.; Dekeyster, E.; Stalmans, I.; Moons, L. MMPs in the trabecular meshwork: Promising targets for future glaucoma therapies? Investig. Ophthalmol. Vis. Sci. 2013, 54, 7756–7763. [Google Scholar] [CrossRef] [PubMed]
  166. van Setten, G.B.; Tervo, T.; Viinikka, L.; Perheentupa, J.; Tarkkanen, A. Epidermal growth factor in human tear fluid: A minireview. Int. Ophthalmol. 1991, 15, 359–362. [Google Scholar] [CrossRef] [PubMed]
  167. Peterson, J.L.; Ceresa, B.P. Epidermal growth factor receptor expression in the corneal epithelium. Cells 2021, 10, 2409. [Google Scholar] [CrossRef] [PubMed]
  168. Szabo, S.; Vincze, A. Growth factors in ulcer healing: Lessons from recent studies. J. Physiol. 2000, 94, 77–81. [Google Scholar] [CrossRef] [PubMed]
  169. Szabo, S.; Deng, X.; Tolstanova, G.; Khomenko, T.; Paunovic, B.; Chen, L.; Jadus, M.; Sandor, Z. Angiogenic and anti-angiogenic therapy for gastrointestinal ulcers: New challenges for rational therapeutic predictions and drug design. Curr. Pharm. Des. 2011, 17, 1633–1642. [Google Scholar] [CrossRef] [PubMed]
  170. Szabo, S.; Gombos, Z.; Sandor, Z. Growth factors in gastrointestinal diseases. BioDrugs 1999, 12, 27–41. [Google Scholar] [CrossRef] [PubMed]
  171. Elliott, J.H. Epidermal growth factor: In vivo ocular studies. Trans. Am. Ophthalmol. Soc. 1980, 78, 629–656. [Google Scholar] [PubMed]
  172. Dartt, D.A. Interaction of EGF family growth factors and neurotransmitters in regulating lacrimal gland secretion. Exp. Eye Res. 2004, 78, 337–345. [Google Scholar] [CrossRef] [PubMed]
  173. Tepavcevic, V.; Hodges, R.R.; Zoukhri, D.; Dartt, D.A. Signal transduction pathways used by EGF to stimulate protein secretion in rat lacrimal gland. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1075–1081. [Google Scholar] [CrossRef] [PubMed]
  174. Kim, M.J.; Jun, R.M.; Kim, W.K.; Hann, H.J.; Chong, Y.H.; Park, H.Y.; Chung, J.H. Optimal concentration of human epidermal growth factor (hEGF) for epithelial healing in experimental corneal alkali wounds. Curr. Eye Res. 2001, 22, 272–279. [Google Scholar] [CrossRef] [PubMed]
  175. Ho, P.C.; Elliott, J.H. Kinetics of corneal epithelial regeneration. II. Epidermal growth factor and topical corticosteroids. Investig. Ophthalmol. 1975, 14, 630–633. [Google Scholar] [PubMed]
  176. Ho, P.C.; Davis, W.H.; Elliott, J.H.; Cohen, S. Kinetics of corneal epithelial regeneration and epidermal growth factor. Investig. Ophthalmol. 1974, 13, 804–809. [Google Scholar] [PubMed]
  177. Lim, M.; Goldstein, M.; Tuli, S.; Schultz, G. Growth factor, cytokine and protease interactions during corneal wound healing. Ocul. Surf. 2003, 2, 253–265. [Google Scholar] [CrossRef]
  178. Tripathi, R.; Raja, S.; Tripathi, B. Prospects for epidermal growth factor in the management of corneal disorders. Surv. Ophthalmol. 1990, 34, 457–462. [Google Scholar] [CrossRef]
  179. Sheardown, H.; Wedge, C.; Chou, L.; Apel, R.; Rootman, D.; Cheng, Y. Continuous epidermal growth factor delivery in corneal epithelial wound healing. Investig. Ophthalmol. Vis. Sci. 1993, 34, 3593–3600. [Google Scholar]
  180. Kirschner, S.; Brazzell, R.; Stern, M.; Baird, L. The use of topical epidermal growth factor for the treatment of nonhealing corneal erosions in dogs. J. Am. Anim. Hosp. Assoc. 1991, 27, 449–452. [Google Scholar]
  181. Daniele, S.; Frati, L.; Santoni, G. The effect of epidermal growth factor (EGF) on the corneal epithelium in humans. Graefes Arch. Clin. Exp. Ophthalmol. 1979, 210, 159–165. [Google Scholar] [CrossRef]
  182. Ganem, S.; Mondon, H.; De Felice, G.P. Local treatment by epidermal growth factor after epikeratoplasty: A double-blind clinical trial. J. Fr. Ophtalmol. 1992, 15, 443–447. [Google Scholar] [PubMed]
  183. Scardovi, C.; De Felice, G.; Gazzaniga, A. Epidermal growth factor in the topical treatment of traumatic corneal ulcers. Ophthalmologica 1993, 206, 119–124. [Google Scholar] [CrossRef]
  184. Dellaert, M.; Casey, T.; Wiffen, S.; Gordon, J.; Johnson, P.; Geerards, A.J.; Rijneveld, W.J.; Remeijer, L.; Mulder, P.G.; Beekhuis, W.H. Influence of topical epidermal growth factor on postkeratoplasty re-epithelialization. Br. J. Ophthalmol. 1997, 81, 391–395. [Google Scholar] [CrossRef][Green Version]
  185. Calel, B.; Fagerholm, P. Human epidermal growth factor: The influence on the healing of surgically closed corneal wounds. Acta Ophthalmol. Scand. Suppl. 1987, 182, 58–61. [Google Scholar] [CrossRef]
  186. Nezu, E.; Ohashi, Y.; Kinoshita, S.; Manabe, R. Recombinant human epidermal growth factor and corneal neovascularization. Jpn. J. Ophthalmol. 1992, 36, 401–406. [Google Scholar] [PubMed]
  187. Taniguchi, E.; Nagae, Y.; Watanabe, H.; Ohashi, Y.; Kinoshita, S.; Manabe, R. The effect of recombinant epidermal growth factor in corneal angiogenesis. Nippon. Ganka Gakkai Zasshi 1991, 95, 52–58. [Google Scholar] [PubMed]
  188. Burling, K.; Seguin, M.A.; Marsh, P.; Brinkman, K.; Madigan, J.; Thurmond, M.; Moon-Massat, P.; Mannis, M.; Murphy, C.J. Effect of topical administration of epidermal growth factor on healing of corneal epithelial defects in horses. Am. J. Vet. Res. 2000, 61, 1150–1155. [Google Scholar] [CrossRef] [PubMed]
  189. Bikbov, M.M.; Khalimov, T.A.; Cerrada-Gimenez, M.; Ragauskas, S.; Kalesnykas, G.; Jonas, J.B. Compatibility of intravitreally applied epidermal growth factor and amphiregulin. Int. Ophthalmol. 2021, 41, 2053–2063. [Google Scholar] [CrossRef] [PubMed]
  190. Fernández-Albarral, J.A.; Ramírez, A.I.; de Hoz, R.; Matamoros, J.A.; Salobrar-García, E.; Elvira-Hurtado, L.; López-Cuenca, I.; Sánchez-Puebla, L.; Salazar, J.J.; Ramírez, J.M. Glaucoma: From pathogenic mechanisms to retinal glial cell response to damage. Front. Cell Neurosci. 2024, 18, 1354569. [Google Scholar] [CrossRef] [PubMed]
  191. Park, J.W.; Hwang, S.R.; Yoon, I.S. Advanced growth factor delivery systems in wound management and skin regeneration. Molecules 2017, 22, 1259. [Google Scholar] [CrossRef] [PubMed]
  192. Kim, H.; Kong, W.H.; Seong, K.Y.; Sung, D.K.; Jeong, H.; Kim, J.K.; Yang, S.Y.; Hahn, S.K. Hyaluronate–epidermal growth factor conjugate for skin wound healing and regeneration. Biomacromolecules 2016, 17, 3694–3705. [Google Scholar] [CrossRef] [PubMed]
  193. Son, S.U.; Jang, S.; Choi, Y.; Park, M.; Son, H.Y.; Huh, Y.M.; Yeom, S.J.; Ham, M.S.; Lee, D.K.; Kim, H.N.; et al. Distinctive nanogels as high-efficiency transdermal carriers for skin wound healing. J. Biomed. Nanotechnol. 2020, 16, 304–314. [Google Scholar] [CrossRef] [PubMed]
  194. Legrand, J.M.D.; Martino, M.M. Growth factor and cytokine delivery systems for wound healing. Cold Spring Harb. Perspect. Biol. 2022, 14, a041234. [Google Scholar] [CrossRef]
  195. Sarıkaya, B.; Yumuşak, N.; Yigin, A.; Sipahioğlu, S.; Yavuz, Ü.; Altay, M.A. Comparison of the effects of human recombinant epidermal growth factor and platelet-rich plasma on healing of rabbit patellar tendon. Eklem Hastalik Cerrahisi 2017, 28, 92–99. [Google Scholar] [CrossRef] [PubMed]
  196. Gülcü, A.; Turhan, E.; Aslan, A.; Çaylı, S.; Alimoğulları, E.; Akcan, G. Investigation of the effect of human recombinant epidermal growth factor on rotator cuff healing: An experimental model. Rev. Bras. Ortop. 2022, 58, 271–278. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  197. Liu, Y.; Liu, Y.; Deng, J.; Li, W.; Nie, X. Fibroblast Growth Factor in Diabetic Foot Ulcer: Progress and Therapeutic Prospects. Front. Endocrinol. 2021, 12, 744868. [Google Scholar] [CrossRef]
  198. Li, L.; Wang, H.; Pang, S.; Wang, L.; Fan, Z.; Ma, C.; Yang, S.; Banda, J.; Hui, Q.; Lv, F.; et al. rhFGF-21 Accelerates Corneal Epithelial Wound Healing through the Attenuation of Oxidative Stress and Inflammatory Mediators in Diabetic Mice. J. Biol. Chem. 2023, 299, 105127. [Google Scholar] [CrossRef]
  199. Ergen, K.S.; Subaşı, S.; Rencber, S.F.; Duruksu, G.; Yazır, Y. Evaluation of Clinical and Histological Effects of KGF-2 and NGF on Corneal Wound Healing in an Experimental Alkali Burn Rabbit Model. Exp. Eye Res. 2022, 223, 109190. [Google Scholar] [CrossRef]
  200. Xiang, X.; Lin, W.; Guan, X.; Zhou, B.; Yuan, Y.; Silva, D.; Wang, Y. Corneal Ulcer Development Due to Sintilimab-Anlotinib Combination Therapy-Induced Dry Eye: A Case Report. Transl. Cancer Res. 2024, 13, 2571–2579. [Google Scholar] [CrossRef]
  201. Yamakawa, S.; Hayashida, K. Advances in Surgical Applications of Growth Factors for Wound Healing. Burn. Trauma 2019, 7, 10. [Google Scholar] [CrossRef] [PubMed]
  202. Scalinci, S.Z.; Scorolli, L.; Meduri, A.; Grenga, P.L.; Corradetti, G.; Metrangolo, C. Effect of Basic Fibroblast Growth Factor and Cytochrome C Peroxidase Combination in Transgenic Mice Corneal Epithelial Healing Process after Excimer Laser Photoablation. Clin. Ophthalmol. 2011, 5, 215–221. [Google Scholar] [CrossRef]
  203. Yan, L.; Wu, W.; Wang, Z.; Li, C.; Lu, X.; Duan, H.; Zhou, J.; Wang, X.; Wan, P.; Song, Y.; et al. Comparative Study of the Effects of Recombinant Human Epidermal Growth Factor and Basic Fibroblast Growth Factor on Corneal Epithelial Wound Healing and Neovascularization In Vivo and In Vitro. Ophthalmic Res. 2013, 49, 150–160. [Google Scholar] [CrossRef]
  204. Yang, Y.; Zhong, J.; Cui, D.; Jensen, L.D. Up-to-Date Molecular Medicine Strategies for Management of Ocular Surface Neovascularization. Adv. Drug Deliv. Rev. 2023, 201, 115084. [Google Scholar] [CrossRef]
  205. Chang, L.K.; Garcia-Cardena, G.; Farnebo, F.; Fannon, M.; Chen, E.J.; Butterfield, C.; Moses, M.A.; Mulligan, R.C.; Folkman, J.; Kaipanen, A. Dose-Dependent Response of FGF-2 for Lymphangiogenesis. Proc. Natl. Acad. Sci. USA 2004, 101, 11658–11663. [Google Scholar] [CrossRef]
  206. Quian, J.; Yu, J.; Zhu, X.; Liang, S. MiR-335 Promotes Corneal Neovascularization by Targeting EGFR. BMC Ophthalmol. 2022, 22, 267. [Google Scholar] [CrossRef] [PubMed]
  207. Gurung, H.R.; Carr, M.M.; Bryant, K.; Chucair-Elliott, A.J.; Carr, D.J.J. Fibroblast Growth Factor-2 Drives and Maintains Progressive Corneal Neovascularization Following HSV-1 Infection. Mucosal Immunol. 2017, 11, 172–185. [Google Scholar] [CrossRef] [PubMed]
  208. Ritchey, E.R.; Zelinka, C.P.; Tang, J.; Liu, J.; Fischer, A.J. The Combination of IGF1 and FGF2 and the Induction of Excessive Ocular Growth and Extreme Myopia. Exp. Eye Res. 2012, 99, 1–16. [Google Scholar] [CrossRef] [PubMed]
  209. Fujikawa, Y.; Nishimura, A.; Senga, Y.; Sudo, A. The Effects of Fibroblast Growth Factor-2 (FGF-2) on Achilles Tendon Injury. Foot Ankle Orthop. 2023, 8, 2473011423S00202. [Google Scholar] [CrossRef]
  210. Turgut, N.; Cengiz Çallıoğlu, F.; Bayraktar, A.; Savran, M.; Aşcı, H.; Gülle, K.; Ünal, M. FGF-2 Enriched Nanofiber Scaffold for Advancing Achilles Tendon Healing: A Comparative Experimental Investigation. Front. Surg. 2024, 11, 1424734. [Google Scholar] [CrossRef]
  211. Ali, J.; Pulatkan, A.; Kara, D.; Tezgel, O.; Misir, A.; Ucan, V.; Bozdag, E.; Yildirim, A.N.; Yildiz, F.; Tuncay, I.; et al. Fibroblast Growth Factor Soaked Collagen Membrane Shows No Biomechanical or Histological Advantages in the Treatment of Chronic Rotator Cuff Tears in a Rabbit Model. Arthroscopy 2024, 40, 683–691. [Google Scholar] [CrossRef]
  212. Zhou, Z.; Song, W.; Zhang, G.; Zhan, S.; Cai, Z.; Yu, W.; He, Y. The Recombinant Human Fibroblast Growth Factor-18 (Sprifermin) Improves Tendon-to-Bone Healing by Promoting Chondrogenesis in a Rat Rotator Cuff Repair Model. J. Shoulder Elb. Surg. 2022, 31, 1617–1627. [Google Scholar] [CrossRef]
  213. Halper, J. Advances in the use of growth factors for treatment of disorders of soft tissues. Adv. Exp. Med. Biol. 2014, 802, 59–76. [Google Scholar] [CrossRef] [PubMed]
  214. Yonemitsu, R.; Tokunaga, T.; Shukunami, C.; Ideo, K.; Arimura, H.; Karasugi, T.; Nakamura, E.; Ide, J.; Hiraki, Y.; Mizuta, H. Fibroblast Growth Factor 2 Enhances Tendon-to-Bone Healing in a Rat Rotator Cuff Repair of Chronic Tears. Am. J. Sports Med. 2019, 47, 1701–1712. [Google Scholar] [CrossRef]
  215. Tang, J.B.; Wu, Y.F.; Cao, Y.; Chen, C.H.; Zhou, Y.L.; Avanessian, B.; Shimada, M.; Wang, X.T.; Liu, P.Y. Basic FGF or VEGF Gene Therapy Corrects Insufficiency in the Intrinsic Healing Capacity of Tendons. Sci. Rep. 2016, 6, 20643. [Google Scholar] [CrossRef] [PubMed]
  216. Roberts, J.H.; Halper, J. Growth Factor Roles in Soft Tissue Physiology and Pathophysiology. Adv. Exp. Med. Biol. 2021, 1348, 139–159. [Google Scholar] [CrossRef]
  217. Blanco-Mezquita, T.; Martinez-Garcia, C.; Proença, R.; Zieske, J.D.; Bonini, S.; Lambiase, A.; Merayo-Lloves, J. Nerve Growth Factor Promotes Corneal Epithelial Migration by Enhancing Expression of Matrix Metalloprotease-9. Investig. Ophthalmol. Vis. Sci. 2013, 54, 3880–3890. [Google Scholar] [CrossRef]
  218. Aloe, L. Nerve growth factor, human skin ulcers and vascularization: Our experience. Prog. Brain Res. 2004, 146, 515–522. [Google Scholar] [CrossRef] [PubMed]
  219. Lambiase, A.; Manni, L.; Rama, P.; Bonini, S. Clinical application of nerve growth factor on human corneal ulcer. Arch. Ital. Biol. 2003, 141, 141–148. [Google Scholar] [PubMed]
  220. Lambiase, A.; Coassin, M.; Sposato, V.; Micera, A.; Sacchetti, M.; Bonini, S.; Aloe, L. NGF topical application in patients with corneal ulcer does not generate circulating NGF antibodies. Pharmacol. Res. 2007, 56, 65–69. [Google Scholar] [CrossRef] [PubMed]
  221. Bruscolini, A.; Marenco, M.; Albanese, G.M.; Lambiase, A.; Sacchetti, M. Long-term clinical efficacy of topical treatment with recombinant human nerve growth factor in neurotrophic keratopathy: A novel cure for a rare degenerative corneal disease? Orphanet J. Rare Dis. 2022, 17, 57. [Google Scholar] [CrossRef] [PubMed]
  222. Roszkowska, A.M.; Spinella, R.; Calderone, A.; Sindoni, M.; Wowra, B.H.; Kozak, M.; Sajak-Hydzik, K.; Aliò, J. The use of rh-NGF in the management of neurotrophic keratopathy. Front. Ophthalmol. 2024, 4, 1408587. [Google Scholar] [CrossRef] [PubMed]
  223. Kahuam-López, N.; Hosseini, A.; Ling, J.Y.M.; Chiang, J.; Iovieno, A.; Yeung, S.N. The Role of Nerve Growth Factor on the Ocular Surface: A Review of the Current Experimental Research and Clinical Practices. Int. J. Mol. Sci. 2025, 26, 6012. [Google Scholar] [CrossRef] [PubMed]
  224. Liu, Z.; Wu, H.; Huang, S. Role of NGF and its receptors in wound healing (Review). Exp. Ther. Med. 2021, 21, 599. [Google Scholar] [CrossRef] [PubMed]
  225. Lambiase, A.; Centofanti, M.; Micera, A.; Manni, G.L.; Mattei, E.; De Gregorio, A.; de Feo, G.; Bucci, M.G.; Aloe, L. Nerve growth factor (NGF) reduces and NGF antibody exacerbates retinal damage induced in rabbit by experimental ocular hypertension. Graefes Arch. Clin. Exp. Ophthalmol. 1997, 235, 780–785. [Google Scholar] [CrossRef] [PubMed]
  226. Colafrancesco, V.; Parisi, V.; Sposato, V.; Rossi, S.; Russo, M.A.; Coassin, M.; Lambiase, A.; Aloe, L. Ocular application of nerve growth factor protects degenerating retinal ganglion cells in a rat model of glaucoma. J. Glaucoma 2011, 20, 100–108. [Google Scholar] [CrossRef] [PubMed]
  227. Lambiase, A.; Aloe, L.; Centofanti, M.; Parisi, V.; Nair Báo, S.; Mantelli, F.; Colafrancesco, V.; Manni, G.; Bucci, M.G.; Bonini, S.; et al. Experimental and clinical evidence of neuroprotection by nerve growth factor eye drops: Implications for glaucoma. Proc. Natl. Acad. Sci. USA 2009, 106, 13469–13474. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  228. Guo, L.; Davis, B.M.; Ravindran, N.; Galvao, J.; Kapoor, N.; Haamedi, N.; Shamsher, E.; Luong, V.; Fico, E.; Cordeiro, M.F. Topical recombinant human nerve growth factor (rh-NGF) is neuroprotective to retinal ganglion cells by targeting secondary degeneration. Sci. Rep. 2020, 10, 60427. [Google Scholar] [CrossRef] [PubMed]
  229. Beykin, G.; Stell, L.; Halim, M.S.; Nuñez, M.; Popova, L.; Nguyen, B.T.; Groth, S.L.; Dennis, A.; Li, Z.; Atkins, M.; et al. Phase 1b randomized controlled study of short-course topical recombinant human nerve growth factor (rhNGF) for neuroenhancement in glaucoma: Safety, tolerability, and efficacy measure outcomes. Am. J. Ophthalmol. 2022, 234, 223–234. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  230. Sposato, V.; Bucci, M.G.; Coassin, M.; Russo, M.A.; Lambiase, A.; Aloe, L. Reduced NGF level and TrkA protein and TrkA gene expression in the optic nerve of rats with experimentally induced glaucoma. Neurosci. Lett. 2008, 446, 20–24. [Google Scholar] [CrossRef] [PubMed]
  231. Mallone, F.; Sacchetti, M.; Bruscolini, A.; Scuderi, L.; Marenco, M.; Lambiase, A. Neurotrophic Factors in Glaucoma and Innovative Delivery Systems. Appl. Sci. 2020, 10, 9015. [Google Scholar] [CrossRef]
  232. Cellini, M.; Bendo, E.; Bravetti, G.O.; Campos, E.C. The use of nerve growth factor in surgical wound healing of the cornea. Ophthalmic Res. 2006, 38, 177–181. [Google Scholar] [CrossRef] [PubMed]
  233. Cherief, M.; Xu, J.; Li, Z.; Tower, R.J.; Ramesh, S.; Qin, Q.; Gomez-Salazar, M.; Yea, J.H.; Lee, S.; Negri, S.; et al. TrkA-mediated sensory innervation of injured mouse tendon supports tendon sheath progenitor cell expansion and tendon repair. Sci. Transl. Med. 2023, 15, eade4619. [Google Scholar] [CrossRef] [PubMed]
  234. Mammoto, T.; Seerattan, R.A.; Paulson, K.D.; Leonard, C.A.; Bray, R.C.; Salo, P.T. Nerve growth factor improves ligament healing. J. Orthop. Res. 2008, 26, 957–964. [Google Scholar] [CrossRef] [PubMed]
  235. Izumi, K.; Kurosaka, D.; Iwata, T.; Oguchi, Y.; Tanaka, Y.; Mashima, Y.; Tsubota, K. Involvement of insulin-like growth factor-I and insulin-like growth factor binding protein-3 in corneal fibroblasts during corneal wound healing. Investig. Ophthalmol. Vis. Sci. 2006, 47, 591–598. [Google Scholar] [CrossRef] [PubMed]
  236. Stuard, W.L.; Titone, R.; Robertson, D.M. The IGF/Insulin-IGFBP axis in corneal development, wound healing, and disease. Front. Endocrinol. 2020, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  237. Peterson, C.; Chandler, H.L. Insulin facilitates corneal wound healing in the diabetic environment through the RTK-PI3K/Akt/mTOR axis in vitro. Mol. Cell. Endocrinol. 2022, 548, 111611. [Google Scholar] [CrossRef] [PubMed]
  238. Chen, S.; Song, W.; Cheng, Y.; Gao, Y.; Xie, L.; Huang, M.; Yan, X. Insulin eye drops improve corneal wound healing in STZ-induced diabetic mice by regulating corneal inflammation and neuropeptide release. BMC Ophthalmol. 2024, 24, 155. [Google Scholar] [CrossRef] [PubMed]
  239. Bastion, M.L.; Ling, K.P. Topical insulin for healing of diabetic epithelial defects?: A retrospective review of corneal debridement during vitreoretinal surgery in Malaysian patients. Med. J. Malays. 2013, 68, 208–216. [Google Scholar] [PubMed]
  240. Tong, C.M.; Iovieno, A.; Yeung, S.N. Topical insulin for neurotrophic corneal ulcers. Can. J. Ophthalmol. 2020, 55, e170–e172. [Google Scholar] [CrossRef] [PubMed]
  241. Leong, C.Y.; Naffi, A.A.; Halim, W.H.W.A.; Bastion, M.L.C. Usage of topical insulin for the treatment of diabetic keratopathy, including corneal epithelial defects. World J. Diabetes 2023, 14, 930–938. [Google Scholar] [CrossRef] [PubMed]
  242. Dasrilsyah, A.M.; Halim, W.H.W.A.; Mustapha, M.; Tang, S.F.; Kaur, B.; Ong, E.Y.; Bastion, M.L.C. Randomized clinical trial of topical insulin versus artificial tears for healing rates of iatrogenic corneal epithelial defects induced during vitreoretinal surgery in diabetics. Cornea 2023, 42, 1395–1403. [Google Scholar] [CrossRef] [PubMed]
  243. Abdi, P.; Ghaffari, R.; Azad, N.; Alshaheeb, A.; Latifi, G.; Soltani Shahgoli, S.; Fakhredin, H. Topical insulin for refractory persistent corneal epithelial defects. Sci. Rep. 2024, 14, 12459. [Google Scholar] [CrossRef] [PubMed]
  244. Díaz-Valle, D.; Burgos-Blasco, B.; Gegúndez-Fernández, J.A.; García-Caride, S.; Puebla-García, V.; Peña-Urbina, P.; Benítez-del-Castillo, J.M. Topical insulin for refractory persistent corneal epithelial defects. Eur. J. Ophthalmol. 2021, 31, 2280–2286. [Google Scholar] [CrossRef] [PubMed]
  245. Krolo, I.; Behaegel, J.; Termote, K.; de Bruyn, B.; De Schepper, M.; Oellerich, S.; Ní Dhubhghaill, S. The role of topical insulin in ocular surface restoration: A review. Surv. Ophthalmol. 2024, 69, 805–817. [Google Scholar] [CrossRef] [PubMed]
  246. Yang, S.; Zhang, Y.; Zhang, Z.; Dan, J.; Zhou, Q.; Wang, X.; Li, W.; Zhou, L.; Yang, L.; Xie, L. Insulin promotes corneal nerve repair and wound healing in type 1 diabetic mice by enhancing Wnt/β-catenin signaling. Am. J. Pathol. 2020, 190, 2237–2250. [Google Scholar] [CrossRef] [PubMed]
  247. Zhang, Y.; Du, Y.; Zhou, S.; Liu, Z.; Li, P.; Du, Z. Topical application of insulin encapsulated by chitosan-modified PLGA nanoparticles to alleviate alkali burn-induced corneal neovascularization. Nanoscale 2025, 17, 12323–12339. [Google Scholar] [CrossRef] [PubMed]
  248. Yu, T.; Gao, M.; Yang, P.; Pei, Q.; Liu, D.; Wang, D.; Zhang, X.; Liu, Y. Topical insulin accelerates cutaneous wound healing in insulin-resistant diabetic rats. Am. J. Transl. Res. 2017, 9, 4682–4693. [Google Scholar] [PubMed]
  249. Durgam, S.S.; Altmann, N.N.; Coughlin, H.E.; Rollins, A.; Hostnik, L.D. Insulin enhances the in vitro osteogenic capacity of flexor tendon-derived progenitor cells. Stem Cells Int. 2019, 2019, 1602751. [Google Scholar] [CrossRef] [PubMed]
  250. Fai, S.; Ahem, A.; Mustapha, M.; Noh, U.K.M.; Bastion, M.L.C. Randomized controlled trial of topical insulin for healing corneal epithelial defects induced during vitreoretinal surgery in diabetics. Asia Pac. J. Ophthalmol. 2017, 6, 418–424. [Google Scholar] [CrossRef] [PubMed]
  251. Zagon, I.S.; Klocek, M.S.; Sassani, J.W.; McLaughlin, P.J. Use of topical insulin to normalize corneal epithelial healing in diabetes mellitus. Arch. Ophthalmol. 2007, 125, 1082–1088. [Google Scholar] [CrossRef] [PubMed]
  252. Zhao, Y.X.; Chen, X.W. Diabetes and risk of glaucoma: Systematic review and a meta-analysis of prospective cohort studies. Int. J. Ophthalmol. 2017, 10, 1430–1435. [Google Scholar] [CrossRef]
  253. Nakamura, M.; Ofuji, K.; Chikama, T.; Nishida, T. Combined effects of substance P and insulin-like growth factor-1 on corneal epithelial wound closure of rabbit in vivo. Curr. Eye Res. 1997, 16, 275–278. [Google Scholar] [CrossRef] [PubMed]
  254. Nishida, T.; Nakamura, M.; Ofuji, K.; Reid, T.W.; Mannis, M.J.; Murphy, C.J. Synergistic effects of substance P with insulin-like growth factor-1 on epithelial migration of the cornea. J. Cell. Physiol. 1996, 169, 159–166. [Google Scholar] [CrossRef] [PubMed]
  255. van Beijnum, J.R.; Pieters, W.; Nowak-Sliwinska, P.; Griffioen, A.W. Insulin-like growth factor axis targeting in cancer and tumour angiogenesis—The missing link. Biol. Rev. 2017, 92, 1755–1768. [Google Scholar] [CrossRef] [PubMed]
  256. Chen, Y.; Zhu, Y.; Song, S.; Hu, Y. Contribution Of Insulin-Like Growth Factor-1 To Tendon Repair (Review). Int. J. Mol. Med. 2025, 56, 195. [Google Scholar] [CrossRef] [PubMed]
  257. Miescher, I.; Rieber, J.; Calcagni, M.; Buschmann, J. In vitro and in vivo effects of IGF-1 delivery strategies on tendon healing: A review. Int. J. Mol. Sci. 2023, 24, 2370. [Google Scholar] [CrossRef] [PubMed]
  258. Kurtz, C.A.; Loebig, T.G.; Anderson, D.D.; DeMeo, P.J.; Campbell, P.G. Insulin-like growth factor I accelerates functional recovery from Achilles tendon injury in a rat model. Am. J. Sports Med. 1999, 27, 363–369. [Google Scholar] [CrossRef] [PubMed]
  259. Li, J.; Weber, E.; Guth-Gundel, S.; Schuleit, M.; Kuttler, A.; Halleux, C.; Accart, N.; Doelemeyer, A.; Basler, A.; Tigani, B.; et al. Tough composite hydrogels with high loading and local release of biological drugs. Adv. Healthc. Mater. 2018, 7, 1701393. [Google Scholar] [CrossRef] [PubMed]
  260. Dahlgren, L.A.; van der Meulen, M.C.; Bertram, J.E.; Starrak, G.S.; Nixon, A.J. Insulin-like growth factor-I improves cellular and molecular aspects of healing in a collagenase-induced model of flexor tendinitis. J. Orthop. Res. 2002, 20, 910–919. [Google Scholar] [CrossRef] [PubMed]
  261. Witte, T.H.; Yeager, A.E.; Nixon, A.J. Intralesional injection of insulin-like growth factor-I for treatment of superficial digital flexor tendonitis in Thoroughbred racehorses: 40 cases (2000–2004). J. Am. Vet. Med. Assoc. 2011, 239, 992–997. [Google Scholar] [CrossRef] [PubMed]
  262. Hansen, M.; Boesen, A.; Holm, L.; Flyvbjerg, A.; Langberg, H.; Kjaer, M. Local administration of insulin-like growth factor-I (IGF-I) stimulates tendon collagen synthesis in humans. Scand. J. Med. Sci. Sports 2013, 23, 614–619. [Google Scholar] [CrossRef] [PubMed]
  263. Olesen, J.L.; Hansen, M.; Turtumoygard, I.F.; Hoffner, R.; Schjerling, P.; Christensen, J.; Mencodias, C.L.; Magnusson, P.S.; Kjaer, M. No treatment benefits of local administration of insulin-like growth factor-1 in addition to heavy slow resistance training in tendinopathic human patellar tendons: A randomized, double-blind, placebo-controlled trial with 1-year follow-up. Am. J. Sports Med. 2021, 49, 2361–2370. [Google Scholar] [CrossRef] [PubMed]
  264. Boisjoly, H.M.; Beaulieu, A.; Brunette, I.; Bazin, R.; Laughrea, P.A.; Chamberland, G.; Heon, E.; Dubé, I. Topical autologous fibronectin in patients with recurrent corneal epithelial defects. Cornea 1991, 10, 483–488. [Google Scholar] [CrossRef] [PubMed]
  265. Brooks, D.E. Matrix metalloproteinase inhibition in corneal ulceration. Ophthalmol. Clin. N. Am. 2004, 17, 33–43. [Google Scholar] [CrossRef] [PubMed]
  266. Yu, F.-S.; Yin, J.; Xu, K.; Huang, J. Growth factors and corneal epithelial wound healing. Brain Res. Bull. 2009, 81, 229–235. [Google Scholar] [CrossRef] [PubMed]
  267. Pflugfelder, S.C.; Massaro-Giordano, M.; Perez, V.L.; Hamrah, P.; Deng, S.X.; Espandar, L.; Foster, C.S.; Affatato, B.; Ng, D.; Cunningham, D.; et al. Topical recombinant human nerve growth factor (cenegermin) for neurotrophic keratopathy: A multicenter randomized vehicle-controlled pivotal trial. Ophthalmology 2020, 127, 14–26. [Google Scholar] [CrossRef] [PubMed]
  268. Pastor, J.C.; Calonge, M. Epidermal growth factor and corneal wound healing: A multicenter study. Cornea 1992, 11, 311–314. [Google Scholar] [CrossRef] [PubMed]
  269. Cho, Y.W.; Yoo, W.S.; Kim, S.J.; Chung, I.Y.; Seo, S.W.; Yoo, J.M. Efficacy of systemic vitamin C supplementation in reducing corneal opacity resulting from infectious keratitis. Medicine 2014, 93, e125. [Google Scholar] [CrossRef] [PubMed]
  270. Meduri, A.; Scorolli, L.; Scalinci, S.Z.; Grenga, P.L.; Lupo, S.; Rechichi, M.; Meduri, E. Effect of the combination of basic fibroblast growth factor and cysteine on corneal epithelial healing after photorefractive keratectomy in patients affected by myopia. Indian J. Ophthalmol. 2014, 62, 424–428. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  271. Petek, M.; Sikiric, P.; Anic, T.; Buljat, G.; Separovic, J.; Stancic-Rokotov, D.; Seiwerth, S.; Grabarevic, Z.; Rucman, R.; Mikus, D.; et al. Pentadecapeptide BPC 157 attenuates gastric lesions induced by alloxan in rats and mice. J. Physiol. Paris 1999, 93, 207–212. [Google Scholar] [CrossRef] [PubMed]
  272. Seveljević-Jaran, D.; Cuzić, S.; Dominis-Kramarić, M.; Glojnarić, I.; Ivetić, V.; Radosević, S.; Parnham, M.J. Accelerated healing of excisional skin wounds by PL 14736 in alloxan-hyperglycemic rats. Skin Pharmacol. Physiol. 2006, 19, 266–274. [Google Scholar] [CrossRef] [PubMed]
  273. Gwyer, D.; Wragg, N.M.; Wilson, S.L. Gastric pentadecapeptide body protection compound BPC 157 and its role in accelerating musculoskeletal soft tissue healing. Cell Tissue Res. 2019, 377, 153–159. [Google Scholar] [CrossRef] [PubMed]
  274. Li, Z.N.; Yuan, Z.F.; Mu, G.Y.; Hu, M.; Cao, L.J.; Zhang, Y.L.; Ge, M.X. Augmented anti-angiogenesis activity of polysulfated heparin-endostatin and polyethylene glycol-endostatin in alkali burn-induced corneal ulcers in rabbits. Exp. Ther. Med. 2015, 10, 889–894. [Google Scholar] [CrossRef] [PubMed][Green Version]
  275. Yoshida, J.; Wicks, R.T.; Zambrano, A.I.; Tyler, B.M.; Javaherian, K.; Grossman, R.; Daoud, Y.J.; Gehlbach, P.; Brem, H.; Stark, W.J. Inhibition of corneal neovascularization by subconjunctival injection of Fc-Endostatin, a novel inhibitor of angiogenesis. J. Ophthalmol. 2015, 2015, 137136. [Google Scholar] [CrossRef] [PubMed]
  276. Ge, H.Y.; Xiao, N.; Yin, X.L.; Fu, S.B.; Ge, J.Y.; Shi, Y.; Liu, P. Comparison of the antiangiogenic activity of modified RGDRGD-endostatin to endostatin delivered by gene transfer in vivo (rabbit alkaline-burn CNV model). Mol. Vis. 2011, 17, 1918–1928. [Google Scholar] [PubMed]
  277. Zhang, P.; Wu, D.; Ge, J.; Zhu, Z.; Feng, G.; Yue, T.; Lin, J.; Zheng, H. Experimental inhibition of corneal neovascularization by endostatin gene transfection in vivo. Chin. Med. J. 2003, 116, 1869–1874. [Google Scholar] [PubMed]
  278. Lai, L.J.; Xiao, X.; Wu, J.H. Inhibition of corneal neovascularization with endostatin delivered by adeno-associated viral (AAV) vector in a mouse corneal injury model. J. Biomed. Sci. 2007, 14, 313–322. [Google Scholar] [CrossRef] [PubMed]
  279. Campochiaro, P.A.; Lauer, A.K.; Sohn, E.H.; Mir, T.A.; Naylor, S.; Anderton, M.C.; Kelleher, M.; Harrop, R.; Ellis, S.; Mitrophanous, K.A. Lentiviral vector gene transfer of endostatin/angiostatin for macular degeneration (GEM) study. Hum. Gene Ther. 2017, 28, 99–111. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  280. Binley, K.; Widdowson, P.S.; Kelleher, M.; de Belin, J.; Loader, J.; Ferrige, G.; Carlucci, M.; Esapa, M.; Chipchase, D.; Angell-Manning, D.; et al. Safety and biodistribution of an equine infectious anemia virus-based gene therapy, RetinoStat®, for age-related macular degeneration. Hum. Gene Ther. 2012, 23, 980–991. [Google Scholar] [CrossRef] [PubMed]
  281. Kachi, S.; Binley, K.; Yokoi, K.; Umeda, N.; Akiyama, H.; Muramatsu, D.; Iqball, S.; Kan, O.; Naylor, S.; Campochiaro, P.A. Equine infectious anemia viral vector-mediated codelivery of endostatin and angiostatin driven by retinal pigmented epithelium-specific VMD2 promoter inhibits choroidal neovascularization. Hum. Gene Ther. 2009, 20, 31–39. [Google Scholar] [CrossRef] [PubMed]
  282. Balaggan, K.S.; Binley, K.; Esapa, M.; MacLaren, R.E.; Iqball, S.; Duran, Y.; Pearson, R.A.; Kan, O.; Barker, S.E.; Smith, A.J.; et al. EIAV vector-mediated delivery of endostatin or angiostatin inhibits angiogenesis and vascular hyperpermeability in experimental CNV. Gene Ther. 2006, 13, 1153–1165. [Google Scholar] [CrossRef] [PubMed][Green Version]
  283. Seppinen, L.; Sormunen, R.; Soini, Y.; Elamaa, H.; Heljasvaara, R.; Pihlajaniemi, T. Lack of collagen XVIII accelerates cutaneous wound healing, while overexpression of its endostatin domain leads to delayed healing. Matrix Biol. 2008, 27, 535–546. [Google Scholar] [CrossRef] [PubMed]
  284. Bloch, W.; Huggel, K.; Sasaki, T.; Grose, R.; Bugnon, P.; Addicks, K.; Timpl, R.; Werner, S. The angiogenesis inhibitor endostatin impairs blood vessel maturation during wound healing. FASEB J. 2000, 14, 2373–2376. [Google Scholar] [CrossRef] [PubMed]
  285. Pufe, T.; Petersen, W.; Kurz, B.; Tsokos, M.; Tillmann, B.; Mentlein, R. Mechanical factors influence the expression of endostatin—An inhibitor of angiogenesis—In tendons. J. Orthop. Res. 2003, 21, 610–616. [Google Scholar] [CrossRef] [PubMed]
  286. Lambert, V.; Munaut, C.; Carmeliet, P.; Gesrard, R.D.; Declerck, P.J.; Gils, A.; Claes, C.; Foidart, J.M.; Noël, A.; Rakic, J.M. Dose-dependent modulation of choroidal neovascularization by plasminogen activator inhibitor type I: Implications for clinical trials. Investig. Ophthalmol. Vis. Sci. 2003, 44, 2791–2797. [Google Scholar] [CrossRef] [PubMed]
  287. Devy, L.; Blacher, S.; Grignet-Debrus, C.; Bajou, K.; Masson, V.; Gerard, R.D.; Gils, A.; Carmeliet, G.; Carmeliet, P.; Declerck, P.J.; et al. The pro- or antiangiogenic effect of plasminogen activator inhibitor 1 is dose dependent. FASEB J. 2002, 16, 147–154. [Google Scholar] [CrossRef] [PubMed]
  288. Basu, A.; Menicucci, G.; Maestas, J.; Das, A.; McGuire, P. Plasminogen activator inhibitor-1 (PAI-1) facilitates retinal angiogenesis in a model of oxygen-induced retinopathy. Investig. Ophthalmol. Vis. Sci. 2009, 50, 4974–4981. [Google Scholar] [CrossRef] [PubMed]
  289. Berman, M.B. Evidence for a role of the plasminogen activator–plasmin system in corneal ulceration. Investig. Ophthalmol. Vis. Sci. 1980, 19, 1204–1221. [Google Scholar] [PubMed]
  290. Sun, H.; Mi, X.; Gao, N.; Yan, C.; Yu, F.S. Hyperglycemia-suppressed expression of Serpine1 contributes to delayed epithelial wound healing in diabetic mouse corneas. Investig. Ophthalmol. Vis. Sci. 2015, 56, 3383–3392. [Google Scholar] [CrossRef] [PubMed]
  291. Wang, Z.; Sosne, G.; Kurpakus-Wheater, M. Plasminogen activator inhibitor-1 (PAI-1) stimulates human corneal epithelial cell adhesion and migration in vitro. Exp. Eye Res. 2005, 80, 1–8. [Google Scholar] [CrossRef] [PubMed]
  292. Providence, K.M.; Higgins, S.P.; Mullen, A.; Battista, A.; Samarakoon, R.; Higgins, C.E.; Wilkins-Port, C.E.; Higgins, P.J. SERPINE1 (PAI-1) is deposited into keratinocyte migration “trails” and required for optimal monolayer wound repair. Arch. Dermatol. Res. 2008, 300, 303–310. [Google Scholar] [CrossRef] [PubMed]
  293. Dan, J.; Belyea, D.; Gertner, G.; Leshem, I.; Lusky, M.; Miskin, R. Plasminogen activator inhibitor-1 in the aqueous humor of patients with and without glaucoma. Arch. Ophthalmol. 2005, 123, 220–224. [Google Scholar] [CrossRef] [PubMed]
  294. Masos, T.; Dan, J.A.; Miskin, R. Plasminogen activator inhibitor-1 mRNA is localized in the ciliary epithelium of the rodent eye. Investig. Ophthalmol. Vis. Sci. 2000, 41, 1006–1011. [Google Scholar] [PubMed]
  295. Fuchshofer, R.; Welge-Lüssen, U.; Lütjen-Drecoll, E. The effect of TGF-β2 on the human trabecular meshwork extracellular proteolytic system. Exp. Eye Res. 2003, 77, 757–765. [Google Scholar] [CrossRef] [PubMed]
  296. Gerometta, R.; Kumar, S.; Shah, S.; Alvarez, L.; Candia, O.; Danias, J. Reduction of steroid-induced intraocular pressure elevation in sheep by tissue plasminogen activator. Investig. Ophthalmol. Vis. Sci. 2013, 54, 7903–7909. [Google Scholar] [CrossRef] [PubMed]
  297. Freeberg, M.A.T.; Farhat, Y.M.; Easa, A.; Kallenbach, J.G.; Malcolm, D.W.; Buckley, M.R.; Benoit, D.S.W.; Awad, H.A. Serpine1 knockdown enhances MMP activity after flexor tendon injury in mice: Implications for adhesions therapy. Sci. Rep. 2018, 8, 5810. [Google Scholar] [CrossRef] [PubMed]
  298. Farhat, Y.M.; Al-Maliki, A.A.; Easa, A.; O’Keefe, R.J.; Schwarz, E.M.; Awad, H.A. TGF-β1 suppresses plasmin and MMP activity in flexor tendon cells via PAI-1: Implications for scarless flexor tendon repair. J. Cell. Physiol. 2015, 230, 318–326. [Google Scholar] [CrossRef] [PubMed]
  299. Freeberg, M.A.T.; Easa, A.; Lillis, J.A.; Benoit, D.S.W.; van Wijnen, A.J.; Awad, H.A. Transcriptomic analysis of cellular pathways in healing flexor tendons of plasminogen activator inhibitor-1 (PAI-1/Serpine1) null mice. J. Orthop. Res. 2020, 38, 43–58. [Google Scholar] [CrossRef] [PubMed]
  300. Ho, T.C.; Chen, S.L.; Wu, J.Y.; Ho, M.Y.; Chen, L.J.; Hsieh, J.W.; Cheng, H.C.; Tsao, Y.P. PEDF promotes self-renewal of limbal stem cell and accelerates corneal epithelial wound healing. Stem Cells 2013, 31, 1775–1784. [Google Scholar] [CrossRef] [PubMed]
  301. Jin, J.; Ma, J.X.; Guan, M.; Yao, K. Inhibition of chemical cautery-induced corneal neovascularization by topical pigment epithelium-derived factor eyedrops. Cornea 2010, 29, 1055–1061. [Google Scholar] [CrossRef] [PubMed]
  302. Matsui, T.; Nishino, Y.; Maeda, S.; Yamagishi, S. PEDF-derived peptide inhibits corneal angiogenesis by suppressing VEGF expression. Microvasc. Res. 2012, 84, 105–108. [Google Scholar] [CrossRef] [PubMed]
  303. Kuo, C.N.; Yang, L.C.; Yang, C.T.; Lai, C.H.; Chen, M.F.; Chen, C.Y.; Chen, C.H.; Wu, P.C.; Kou, H.K.; Chen, Y.J.; et al. Inhibition of corneal neovascularization with plasmid pigment epithelium-derived factor (p-PEDF) delivered by synthetic amphiphile INTeraction-18 (SAINT-18) vector in an experimental model of rat corneal angiogenesis. Exp. Eye Res. 2009, 89, 678–685. [Google Scholar] [CrossRef] [PubMed]
  304. Yeh, S.I.; Ho, T.C.; Chen, S.L.; Chen, C.P.; Cheng, H.C.; Lan, Y.W.; Hsieh, J.W.; Wang, C.T.; Tsao, Y.P. Pigment epithelial-derived factor peptide regenerated limbus serves as regeneration source for limbal regeneration in rabbit limbal deficiency. Investig. Ophthalmol. Vis. Sci. 2016, 57, 2629–2636. [Google Scholar] [CrossRef] [PubMed]
  305. Rogers, M.E.; Navarro, I.D.; Perkumas, K.M.; Niere, S.M.; Allingham, R.R.; Crosson, C.E.; Stamer, W.D. Pigment epithelium-derived factor decreases outflow facility. Investig. Ophthalmol. Vis. Sci. 2013, 54, 6655–6661. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  306. Yoshida, Y.; Yamagishi, S.; Matsui, T.; Nakamura, K.; Imaizumi, T.; Yoshimura, K.; Yamakawa, R. Increased levels of pigment epithelium-derived factor in the aqueous humor of patients with uveitis. Br. J. Ophthalmol. 2007, 91, 149–150. [Google Scholar] [CrossRef] [PubMed]
  307. Kaeslin, M.A.; Killer, H.E.; Fuhrer, C.A.; Zeleny, N.; Huber, A.R.; Neutzner, A. Changes to the aqueous humor proteome during glaucoma. PLoS ONE 2016, 11, e0165314. [Google Scholar] [CrossRef] [PubMed]
  308. Sharma, S.; Bollinger, K.E.; Kodeboyina, S.K.; Zhi, W.; Patton, J.; Bai, S.; Edwards, B.; Ulrich, L.; Bogorad, D.; Sharma, A. Proteomic alterations in aqueous humor from patients with primary open angle glaucoma. Investig. Ophthalmol. Vis. Sci. 2018, 59, 2635–2643. [Google Scholar] [CrossRef] [PubMed]
  309. Ho, T.C.; Tsai, S.H.; Yeh, S.I.; Chen, S.L.; Tung, K.Y.; Chien, H.Y.; Lu, Y.C.; Huang, C.H.; Tsao, Y.P. PEDF-derived peptide promotes tendon regeneration through its mitogenic effect on tendon stem/progenitor cells. Stem Cell Res. Ther. 2019, 10, 2. [Google Scholar] [CrossRef] [PubMed]
  310. Ho, T.C.; Chiang, Y.P.; Chuang, C.K.; Chen, S.L.; Hsieh, J.W.; Lan, Y.W.; Tsao, Y.P. PEDF-derived peptide promotes skeletal muscle regeneration through its mitogenic effect on muscle progenitor cells. Am. J. Physiol. Cell Physiol. 2015, 309, C159–C168. [Google Scholar] [CrossRef] [PubMed]
  311. Shih, S.C.; Ho, T.C.; Chen, S.L.; Tsao, Y.P. Pigment epithelium-derived factor (PEDF) peptide promotes the expansion of hepatic stem/progenitor cells via ERK and STAT3-dependent signaling. Am. J. Transl. Res. 2017, 9, 1114–1126. [Google Scholar] [PubMed]
  312. Ambati, B.K.; Joussen, A.M.; Ambati, J.; Moromizato, Y.; Guha, C.; Javaherian, K.; Gillies, S.; O’Reilly, M.S.; Adamis, A.P. Angiostatin inhibits and regresses corneal neovascularization. Arch. Ophthalmol. 2002, 120, 1063–1068. [Google Scholar] [CrossRef] [PubMed]
  313. Gabison, E.E.; Chang, J.H.; Hernández-Quintela, E.; Javier, J.; Lu, P.C.; Ye, H.; Kure, T.; Kato, T.; Azar, D.T. Anti-angiogenic role of angiostatin during corneal wound healing. Exp. Eye Res. 2004, 78, 579–589. [Google Scholar] [CrossRef] [PubMed]
  314. Cheng, H.C.; Yeh, S.I.; Tsao, Y.P.; Kuo, P.C. Subconjunctival injection of recombinant AAV-angiostatin ameliorates alkali burn induced corneal angiogenesis. Mol. Vis. 2007, 13, 2344–2352. [Google Scholar] [PubMed]
  315. Chesnokova, N.B.; Aĭsina, R.B.; Mukhametova, L.I.; Pavlenko, T.A.; Gulin, D.A.; Beznos, O.V. Fibrinolysis components and angiogenesis regulation by example of burn-induced corneal neovascularization in rabbits. Vestn. Oftalmol. 2012, 128, 62–65. [Google Scholar] [PubMed]
  316. Helgeland, E.; Pedersen, T.O.; Rashad, A.; Johannessen, A.C.; Mustafa, K.; Rosén, A. Angiostatin-functionalized collagen scaffolds suppress angiogenesis but do not induce chondrogenesis by mesenchymal stromal cells in vivo. J. Oral Sci. 2020, 62, 371–376. [Google Scholar] [CrossRef] [PubMed]
  317. Foulsham, W.; Dohlman, T.H.; Mittal, S.K.; Taketani, Y.; Singh, R.B.; Masli, S.; Dana, R. Thrombospondin-1 in ocular surface health and disease. Ocul. Surf. 2019, 17, 374–383. [Google Scholar] [CrossRef] [PubMed]
  318. Uno, K.; Hayashi, H.; Kuroki, M.; Uchida, H.; Yamauchi, Y.; Kuroki, M.; Oshima, K. Thrombospondin-1 accelerates wound healing of corneal epithelia. Biochem. Biophys. Res. Commun. 2004, 315, 928–934. [Google Scholar] [CrossRef] [PubMed]
  319. Tan, X.; Chen, Y.; Foulsham, W.; Amouzegar, A.; Inomata, T.; Liu, Y.; Chauhan, S.K.; Dana, R. The immunoregulatory role of corneal epithelium-derived thrombospondin-1 in dry eye disease. Ocul. Surf. 2018, 16, 470–477. [Google Scholar] [CrossRef] [PubMed]
  320. Lai, Y.H.; Lee, P.Y.; Lu, C.Y.; Liu, Y.R.; Wang, S.C.; Liu, C.C.; Chang, Y.C.; Chen, Y.H.; Su, C.C.; Li, C.Y.; et al. Thrombospondin 1-induced exosomal proteins attenuate hypoxia-induced paraptosis in corneal epithelial cells and promote wound healing. FASEB J. 2021, 35, e21200. [Google Scholar] [CrossRef] [PubMed]
  321. Haddadin, R.I.; Oh, D.J.; Kang, M.H.; Villarreal, G., Jr.; Kang, J.H.; Jin, R.; Gong, H.; Rhee, D.J. Thrombospondin-1 (TSP1)–null and TSP2-null mice exhibit lower intraocular pressures. Investig. Ophthalmol. Vis. Sci. 2012, 53, 6708–6717. [Google Scholar] [CrossRef] [PubMed]
  322. Fu, H.; Siggs, O.M.; Knight, S.L.; Staffieri, S.E.; Ruddle, J.B.; Birsner, A.E.; Collantes, E.R.; Craig, J.E.; Wiggs, J.L.; D’Amato, R.J. Thrombospondin 1 missense alleles induce extracellular matrix protein aggregation and TM dysfunction in congenital glaucoma. J. Clin. Investig. 2022, 132, e156967. [Google Scholar] [CrossRef] [PubMed]
  323. Kennedy, S.M.; Sheridan, C.; Kearns, V.R.; Bilir, E.K.; Fan, X.; Grierson, I.; Choudhary, A. Thrombospondin-2 is up-regulated by TGFβ2 and increases fibronectin expression in human trabecular meshwork cells. Exp. Eye Res. 2019, 189, 107820. [Google Scholar] [CrossRef] [PubMed]
  324. Murphy-Ullrich, J.E. Thrombospondin-1 signaling through the calreticulin/LDL receptor related protein 1 axis: Functions and possible roles in glaucoma. Front. Cell Dev. Biol. 2022, 10, 898772. [Google Scholar] [CrossRef] [PubMed]
  325. Kyriakides, T.R.; Zhu, Y.H.; Smith, L.T.; Bain, S.D.; Yang, Z.; Lin, M.T.; Danielson, K.G.; Iozzo, R.V.; LaMarca, M.; McKinney, C.E.; et al. Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis. J. Cell Biol. 1998, 140, 419–430. [Google Scholar] [CrossRef] [PubMed]
  326. Streit, M.; Velasco, P.; Riccardi, L.; Spencer, L.; Brown, L.F.; Janes, L.; Lange-Asschenfeldt, B.; Yano, K.; Hawighorst, T.; Iruela-Arispe, M.L.; et al. Thrombospondin-1 suppresses wound healing and granulation tissue formation in the skin of transgenic mice. EMBO J. 2000, 19, 3272–3282. [Google Scholar] [CrossRef] [PubMed]
  327. Kunkemoeller, B.L.; Bancroft, T.; Xing, H.; Morris, A.H.; Luciano, A.K.; Wu, J.; Fernández-Hernando, C.; Kyriakides, T.R. Elevated thrombospondin-2 contributes to delayed wound healing in diabetes. Diabetes 2019, 68, 2016–2023. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  328. Rosini, S.; Pugh, N.; Bonna, A.M.; Hulmes, D.J.S.; Farndale, R.W.; Adams, J.C. Thrombospondin-1 promotes matrix homeostasis by interacting with collagen and lysyl oxidase precursors and collagen cross-linking sites. Sci. Signal. 2018, 11, eaar2566. [Google Scholar] [CrossRef] [PubMed]
  329. Soto-Pantoja, D.R.; Shih, H.B.; Maxhimer, J.B.; Cook, K.L.; Ghosh, A.; Isenberg, J.S.; Roberts, D.D. Thrombospondin-1 and CD47 signaling regulate healing of thermal injury in mice. Matrix Biol. 2014, 37, 25–34. [Google Scholar] [CrossRef] [PubMed]
  330. Moazami, G.; Auran, J.D.; Florakis, G.J.; Wilson, S.E.; Srinivasan, D.B. Interferon treatment of Mooren’s ulcers associated with hepatitis C. Am. J. Ophthalmol. 1995, 119, 365–366. [Google Scholar] [CrossRef] [PubMed]
  331. Minkovitz, J.B.; Pepose, J.S. Topical interferon alpha-2a treatment of herpes simplex keratitis resistant to multiple antiviral medications in an immunosuppressed patient. Cornea 1995, 14, 326–330. [Google Scholar] [CrossRef] [PubMed]
  332. Erdem, U.; Kerimoglu, H.; Cakir Gundogan, F.; Dagli, S. Treatment of Mooren’s ulcer with topical administration of interferon alfa-2a. Ophthalmology 2007, 114, 446–449. [Google Scholar] [CrossRef] [PubMed]
  333. Aaltonen, V.; Alavesa, M.; Pirilä, L.; Vesti, E.; Al-Juhaish, M. Case report: Bilateral Mooren ulcer in association with hepatitis C. BMC Ophthalmol. 2017, 17, 239. [Google Scholar] [CrossRef] [PubMed]
  334. Zarei-Ghanavati, S.; Alizadeh, R.; Deng, S.X. Topical interferon alpha-2b for treatment of noninvasive ocular surface squamous neoplasia with 360° limbal involvement. J. Ophthalmic Vis. Res. 2014, 9, 423–426. [Google Scholar] [CrossRef] [PubMed]
  335. Miller, J.W.; Stinson, W.G.; Folkman, J. Regression of experimental iris neovascularization with systemic alpha-interferon. Ophthalmology 1993, 100, 9–14. [Google Scholar] [CrossRef] [PubMed]
  336. Nguyen, N.; Goldberg, M.; Pico, J.; Kim, W.; Abbott, R.L.; Levy, B. Topical and subcutaneous alpha-interferon fails to suppress corneal neovascularization. Cornea 1995, 14, 147–151. [Google Scholar] [CrossRef] [PubMed]
  337. Bilgin, S. Comparison of the effects of subconjunctival injections of bevacizumab and interferon alpha-2a on corneal angiogenesis in a rat model. Medicina 2018, 54, 16. [Google Scholar] [CrossRef] [PubMed]
  338. Ayaki, M. Development of neovascular glaucoma in the course of interferon alfa therapy for hepatitis type C. Br. J. Ophthalmol. 1994, 78, 238. [Google Scholar] [CrossRef] [PubMed][Green Version]
  339. Kwon, Y.S.; Choe, Y.H.; Chin, H.S. Development of glaucoma in the course of interferon alpha therapy for chronic hepatitis B. Yonsei Med. J. 2001, 42, 134–136. [Google Scholar] [CrossRef] [PubMed]
  340. Rachitskaya, A.V.; Lee, R.K.; Dubovy, S.R.; Schiff, E.R. Combined central retinal vein and central retinal artery occlusions and neovascular glaucoma associated with interferon treatment. Eur. J. Ophthalmol. 2012, 22, 284–287. [Google Scholar] [CrossRef] [PubMed]
  341. Ilyas, F.; Singh, H.; Anand, N.; Ahmed, I.I.K. Intraocular pressure rise in the course of peginterferon alpha-2a, ribavirin, and boceprevir therapy for hepatitis C. Can. J. Ophthalmol. 2015, 50, e112–e114. [Google Scholar] [CrossRef] [PubMed]
  342. Fatemi, M.J.; Shirani, S.; Sobhani, R.; Lebaschi, A.H.; Gharegozlou, M.J.; Bagheri, T.; Pedram, M.; Saberi, M.; Araghi, S.; Fatemi, M.A. Prevention of peritendinous adhesion formation after the flexor tendon surgery in rabbits: A comparative study between use of local interferon-α, interferon-β, and 5-fluorouracil. Ann. Plast. Surg. 2018, 80, 171–175. [Google Scholar] [CrossRef] [PubMed]
  343. Nedelec, B.; Shen, Y.J.; Ghahary, A.; Scott, P.G.; Tredget, E.E. The effect of interferon alpha 2b on the expression of cytoskeletal proteins in an in vitro model of wound contraction. J. Lab. Clin. Med. 1995, 126, 474–484. [Google Scholar] [PubMed]
  344. Sahara, K.; Kucukcelebi, A.; Ko, F.; Phillips, L.; Robson, M. Suppression of in vitro proliferative scar fibroblast contraction by interferon alfa-2b. Wound Repair Regen. 1993, 1, 22–27. [Google Scholar] [CrossRef] [PubMed]
  345. Nedelec, B.; Dodd, C.M.; Scott, P.G.; Ghahary, A.; Tredget, E.E. Effect of interferon-alpha2b on guinea pig wound closure and the expression of cytoskeletal proteins in vivo. Wound Repair Regen. 1998, 6, 202–212. [Google Scholar] [CrossRef] [PubMed]
  346. Ryu, J.S.; Ko, J.H.; Kim, M.K.; Wee, W.R.; Oh, J.Y. Prednisolone induces apoptosis in corneal epithelial cells through the intrinsic pathway. Sci. Rep. 2017, 7, 4135. [Google Scholar] [CrossRef] [PubMed]
  347. Araki-Sasaki, K.; Katsuta, O.; Mano, H.; Nagano, T.; Nakamura, M. The effects of oral and topical corticosteroid in rabbit corneas. BMC Ophthalmol. 2016, 16, 160. [Google Scholar] [CrossRef]
  348. Nakagawa, H.; Koike, N.; Ehara, T.; Hattori, T.; Narimatsu, A.; Kumakura, S.; Goto, H. Corticosteroid eye-drop instillation aggravates the development of Acanthamoeba keratitis in rabbit corneas inoculated with Acanthamoeba and bacteria. Sci. Rep. 2019, 9, 12821. [Google Scholar] [CrossRef]
  349. Cho, C.H.; Lee, S.B. Clinical analysis of microbiologically proven fungal keratitis according to prior topical steroid use: A retrospective study in South Korea. BMC Ophthalmol. 2019, 19, 207. [Google Scholar] [CrossRef]
  350. Knutsson, K.A.; Iovieno, A.; Matuska, S.; Fontana, L.; Rama, P. Topical corticosteroids and fungal keratitis: A review of the literature and case series. J. Clin. Med. 2021, 10, 1178. [Google Scholar] [CrossRef]
  351. Wilhelmus, K.R. Indecision about corticosteroids for bacterial keratitis: An evidence-based update. Ophthalmology 2002, 109, 1021–1028. [Google Scholar] [CrossRef]
  352. Li, W.W.; Casey, R.; Gonzalez, E.M.; Folkman, J. Angiostatic steroids potentiated by sulfated cyclodextrins inhibit corneal neovascularization. Investig. Ophthalmol. Vis. Sci. 1991, 32, 2898–2905. [Google Scholar]
  353. Proia, A.D.; Hirakata, A.; McInnes, J.S.; Scroggs, M.W.; Parikh, I. The effect of angiostatic steroids and beta-cyclodextrin tetradecasulfate on corneal neovascularization in the rat. Exp. Eye Res. 1993, 57, 693–698. [Google Scholar] [CrossRef] [PubMed]
  354. BenEzra, D.; Griffin, B.W.; Maftzir, G.; Sharif, N.A.; Clark, A.F. Topical formulations of novel angiostatic steroids inhibit rabbit corneal neovascularization. Invest. Ophthalmol. Vis. Sci. 1997, 38, 1954–1962. [Google Scholar] [PubMed]
  355. Murata, M.; Shimizu, S.; Horiuchi, S.; Taira, M. Inhibitory effect of triamcinolone acetonide on corneal neovascularization. Graefes Arch. Clin. Exp. Ophthalmol. 2006, 244, 205–209. [Google Scholar] [CrossRef]
  356. Nakao, S.; Hata, Y.; Miura, M.; Noda, K.; Kimura, Y.N.; Kawahara, S.; Kita, T.; Hisatomi, T.; Nakazawa, T.; Jin, Y.; et al. Dexamethasone inhibits interleukin-1β-induced corneal neovascularization: Role of nuclear factor-κB-activated stromal cells in inflammatory angiogenesis. Am. J. Pathol. 2007, 171, 1058–1065. [Google Scholar] [CrossRef]
  357. Hos, D.; Saban, D.R.; Bock, F.; Regenfuss, B.; Onderka, J.; Masli, S.; Cursiefen, C. Suppression of inflammatory corneal lymphangiogenesis by application of topical corticosteroids. Arch. Ophthalmol. 2011, 129, 445–452. [Google Scholar] [CrossRef]
  358. Gordon, D.M.; McLean, J.M.; Koteen, H. Present status of corticotropin; ACTH, cortisone, and hydrocortisone in ophthalmology. Br. J. Ophthalmol. 1953, 37, 85–98. [Google Scholar] [CrossRef] [PubMed][Green Version]
  359. Armaly, M.F. Effect of corticosteroids on intraocular pressure and fluid dynamics. I. The effect of dexamethasone in the normal eye. Arch. Ophthalmol. 1963, 70, 482–491. [Google Scholar] [CrossRef]
  360. Armaly, M.F. Effect of corticosteroids on intraocular pressure and fluid dynamics. II. The effect of dexamethasone in the glaucomatous eye. Arch. Ophthalmol. 1963, 70, 492–499. [Google Scholar] [CrossRef]
  361. Becker, B. Intraocular pressure response to topical corticosteroids. Investig. Ophthalmol. 1965, 4, 198–205. [Google Scholar]
  362. Kitazawa, Y. Increased intraocular pressure induced by corticosteroids. Am. J. Ophthalmol. 1976, 82, 492–495. [Google Scholar] [CrossRef]
  363. Fingert, J.H.; Clark, A.F.; Craig, J.E.; Alward, W.L.; Snibson, G.R.; McLaughlin, M.; Tuttle, L.; Mackey, D.A.; Sheffield, V.C.; Stone, E.M. Evaluation of the myocilin (MYOC) glaucoma gene in monkey and human steroid-induced ocular hypertension. Investig. Ophthalmol. Vis. Sci. 2001, 42, 145–152. [Google Scholar] [PubMed]
  364. Garbe, E.; LeLorier, J.; Boivin, J.F.; Suissa, S. Risk of ocular hypertension or open-angle glaucoma in elderly patients on oral glucocorticoids. Lancet 1997, 350, 979–982. [Google Scholar] [CrossRef] [PubMed]
  365. Kersey, J.P.; Broadway, D.C. Corticosteroid-induced glaucoma: A review of the literature. Eye 2006, 20, 407–416. [Google Scholar] [CrossRef]
  366. Phulke, S.; Kaushik, S.; Kaur, S.; Pandav, S.S. Steroid-induced glaucoma: An avoidable irreversible blindness. J. Curr. Glaucoma Pract. 2017, 11, 67–72. [Google Scholar] [CrossRef]
  367. Wrenn, R.N.; Goldner, J.L.; Markee, J.L. An experimental study of the effect of cortisone on the healing process and tensile strength of tendons. J. Bone Jt. Surg. Am. 1954, 36-A, 588–601. [Google Scholar] [CrossRef] [PubMed]
  368. Phelps, D.; Sonstegard, D.A.; Matthews, L.S. Corticosteroid injection effects on the biomechanical properties of rabbit patellar tendons. Clin. Orthop. Relat. Res. 1974, 100, 345–348. [Google Scholar] [CrossRef] [PubMed]
  369. Matthews, L.S.; Sonstegard, D.A.; Phelps, D.B. A biomechanical study of rabbit patellar tendon: Effects of steroid injection. Am. J. Sports Med. 1974, 2, 349–357. [Google Scholar] [CrossRef]
  370. Kapetanos, G. The effect of the local corticosteroids on the healing and biomechanical properties of the partially injured tendon. Clin. Orthop. Relat. Res. 1982, 163, 170–179. [Google Scholar] [CrossRef] [PubMed]
  371. Tillander, B.; Franzén, L.E.; Karlsson, M.H.; Norlin, R. Effect of steroid injections on the rotator cuff: An experimental study in rats. J. Shoulder Elb. Surg. 1999, 8, 271–274. [Google Scholar] [CrossRef] [PubMed]
  372. Tatari, H.; Kosay, C.; Baran, O.; Ozcan, O.; Ozer, E. Deleterious effects of local corticosteroid injections on the Achilles tendon of rats. Arch. Orthop. Trauma Surg. 2001, 121, 333–337. [Google Scholar] [CrossRef]
  373. Akpinar, S.; Hersekli, M.A.; Demirörs, H.; Tandogan, R.N.; Kayaselçuk, F. Effects of methylprednisolone and betamethasone injections on the rotator cuff: An experimental study in rats. Adv. Ther. 2002, 19, 194–201. [Google Scholar] [CrossRef]
  374. Mikolyzk, D.K.; Wei, A.S.; Tonino, P.; Marra, G.; Williams, D.A.; Himes, R.D.; Wezeman, F.H.; Callaci, J.J. Effect of corticosteroids on the biomechanical strength of rat rotator cuff tendon. J. Bone Jt. Surg. Am. 2009, 91, 1172–1180. [Google Scholar] [CrossRef] [PubMed]
  375. Jost, H.E.; Keenan, A.V.; Keys, D.A.; Myrna, K.E.; Diehl, K.A. Effect of topical non-steroidal anti-inflammatory drugs on healing times and complications in dogs with spontaneous chronic corneal epithelial defects. Vet. Rec. 2022, 190, e1118. [Google Scholar] [CrossRef] [PubMed]
  376. Iwamoto, S.; Koga, T.; Ohba, M.; Okuno, T.; Koike, M.; Murakami, A.; Matsuda, A.; Yokomizo, T. Non-steroidal anti-inflammatory drug delays corneal wound healing by reducing production of 12-hydroxyheptadecatrienoic acid, a ligand for leukotriene B4 receptor 2. Sci. Rep. 2017, 7, 13267. [Google Scholar] [CrossRef] [PubMed]
  377. Lima, T.B.; Ribeiro, A.P.; da Conceição, L.F.; Bandarra, M.; Manrique, W.G.; Laus, J.L. Ketorolac eye drops reduce inflammation and delay re-epithelization in response to corneal alkali burn in rabbits, without affecting iNOS or MMP-9. Arq. Bras. Oftalmol. 2015, 78, 67–72. [Google Scholar] [CrossRef] [PubMed][Green Version]
  378. Miller, D.; Gruenberg, P.; Miller, R.; Bergamini, M.V. Topical flurbiprofen or prednisolone: Effect on corneal wound healing in rabbits. Arch. Ophthalmol. 1981, 99, 681–682. [Google Scholar] [CrossRef] [PubMed]
  379. Schechter, B.A.; Trattler, W. Efficacy and safety of bromfenac for the treatment of corneal ulcer pain. Adv. Ther. 2010, 27, 756–761. [Google Scholar] [CrossRef] [PubMed]
  380. Yani, E.V.; Golikova, V.A. Features of anti-inflammatory therapy of bacterial corneal ulcers. Vestn. Oftalmol. 2020, 136, 41–45. [Google Scholar] [CrossRef] [PubMed]
  381. Cooper, C.A.; Bergamini, M.V.; Leopold, I.H. Use of flurbiprofen to inhibit corneal neovascularization. Arch. Ophthalmol. 1980, 98, 1102–1105. [Google Scholar] [CrossRef] [PubMed]
  382. Duffin, R.M.; Weissmann, B.A.; Glasser, D.B.; Pettit, T.H. Flurbiprofen in the treatment of corneal neovascularization induced by contact lenses. Am. J. Ophthalmol. 1982, 93, 607–614. [Google Scholar] [CrossRef] [PubMed]
  383. Frucht, J.; Zauberman, H. Topical indomethacin: Effect on neovascularisation of the cornea and on prostaglandin E2 levels. Br. J. Ophthalmol. 1984, 68, 656–659. [Google Scholar] [CrossRef] [PubMed]
  384. Haynes, W.L.; Proia, A.D.; Klintworth, G.K. Effect of inhibitors of arachidonic acid metabolism on corneal neovascularization in the rat. Investig. Ophthalmol. Vis. Sci. 1989, 30, 1588–1593. [Google Scholar] [PubMed]
  385. Song, L.; Liu, X.; Chen, N.; Liu, J.; Nie, A.; Li, W. The effect of bromfenac sodium nanopolymer used in anterior segment of the eye on corneal neovascularization. Cell Mol. Biol. 2022, 68, 330–338. [Google Scholar] [CrossRef] [PubMed]
  386. Kashiwagi, K.; Tsukahara, S. Effect of non-steroidal anti-inflammatory ophthalmic solution on intraocular pressure reduction by latanoprost. Br. J. Ophthalmol. 2003, 87, 297–301. [Google Scholar] [CrossRef] [PubMed]
  387. Chiba, T.; Kashiwagi, K.; Chiba, N.; Tsukahara, S. Effect of non-steroidal anti-inflammatory ophthalmic solution on intraocular pressure reduction by latanoprost in patients with primary open angle glaucoma or ocular hypertension. Br. J. Ophthalmol. 2006, 90, 314–317. [Google Scholar] [CrossRef] [PubMed] [PubMed Central][Green Version]
  388. Costagliola, C.; Campa, C.; Perri, P.; Parmeggiani, F.; Romano, M.R.; Incorvaia, C. Topical and oral ketorolac administration increases the intraocular pressure-lowering effect of latanoprost. Curr. Eye Res. 2008, 33, 477–482. [Google Scholar] [CrossRef] [PubMed]
  389. Costagliola, C.; Parmeggiani, F.; Antinozzi, P.P.; Caccavale, A.; Cotticelli, L.; Sebastiani, A. The influence of diclofenac ophthalmic solution on the intraocular pressure-lowering effect of topical 0.5% timolol and 0.005% latanoprost in primary open-angle glaucoma patients. Exp. Eye Res. 2005, 81, 610–615. [Google Scholar] [CrossRef] [PubMed]
  390. Turan-Vural, E.; Torun-Acar, B.; Acar, S. Effect of ketorolac add-on treatment on intra-ocular pressure in glaucoma patients receiving prostaglandin analogues. Ophthalmologica 2012, 227, 205–209. [Google Scholar] [CrossRef] [PubMed]
  391. Lo, K.J.; Ko, Y.C.; Hwang, D.K.; Liu, C.J. The influence of topical non-steroidal anti-inflammatory drugs on the intraocular pressure lowering effect of topical prostaglandin analogues—A systematic review and meta-analysis. PLoS ONE 2020, 15, e0239233. [Google Scholar] [CrossRef] [PubMed]
  392. Cohen, D.B.; Kawamura, S.; Ehteshami, J.R.; Rodeo, S.A. Indomethacin and celecoxib impair rotator cuff tendon-to-bone healing. Am. J. Sports Med. 2006, 34, 362–369. [Google Scholar] [CrossRef] [PubMed]
  393. Ferry, S.T.; Dahners, L.E.; Afshari, H.M.; Weinhold, P.S. The effects of common anti-inflammatory drugs on the healing rat patellar tendon. Am. J. Sports Med. 2007, 35, 1326–1333. [Google Scholar] [CrossRef] [PubMed]
  394. Dimmen, S.; Engebretsen, L.; Nordsletten, L.; Madsen, J.E. Negative effects of parecoxib and indomethacin on tendon healing: An experimental study in rats. Knee Surg. Sports Traumatol. Arthrosc. 2009, 17, 835–839. [Google Scholar] [CrossRef] [PubMed]
  395. Dimmen, S.; Nordsletten, L.; Engebretsen, L.; Steen, H.; Madsen, J.E. The effect of parecoxib and indometacin on tendon-to-bone healing in a bone tunnel: An experimental study in rats. J. Bone Jt. Surg. Br. 2009, 91, 259–263. [Google Scholar] [CrossRef] [PubMed]
  396. Connizzo, B.K.; Yannascoli, S.M.; Tucker, J.J.; Caro, A.C.; Riggin, C.N.; Mauck, R.L.; Soslowsky, L.J.; Steinberg, D.R.; Bernstein, J. The detrimental effects of systemic ibuprofen delivery on tendon healing are time-dependent. Clin. Orthop. Relat. Res. 2014, 472, 2433–2439. [Google Scholar] [CrossRef] [PubMed]
  397. Hill, J.C.; Potter, P. Treatment of Mooren’s ulcer with cyclosporin A: Report of three cases. Br. J. Ophthalmol. 1987, 71, 11–15. [Google Scholar] [CrossRef] [PubMed]
  398. Zierhut, M.; Thiel, H.J.; Weidle, E.G.; Waetjen, R.; Pleyer, U.P. Topical treatment of severe corneal ulcers with cyclosporin A. Graefes. Arch. Clin. Exp. Ophthalmol. 1989, 227, 30–35. [Google Scholar] [CrossRef] [PubMed]
  399. Gottsch, J.D.; Akpek, E.K. Topical cyclosporin stimulates neovascularization in resolving sterile rheumatoid central corneal ulcers. Trans. Am. Ophthalmol. Soc. 2000, 98, 81–90. [Google Scholar] [PubMed]
  400. Zhao, J.C.; Jin, X.Y. Treatment of severe Mooren’s ulcer with cyclosporin A eyedrops. Chin. Med. J. 1992, 105, 406–409. [Google Scholar] [PubMed]
  401. Tandon, R.; Chawla, B.; Verma, K.; Sharma, N.; Titiyal, J.S. Outcome of treatment of Mooren ulcer with topical cyclosporine A 2%. Cornea 2008, 27, 859–861. [Google Scholar] [CrossRef] [PubMed]
  402. Liao, R.; Li, J.; Su, Y.; Tao, Y.; Su, R.; Tan, X. Topical application of 0.05% cyclosporine for the treatment of neurotrophic keratopathy secondary to herpes simplex keratitis. Clin. Ther. 2024, 46, 900–904. [Google Scholar] [CrossRef] [PubMed]
  403. Lipman, R.M.; Epstein, R.J.; Hendricks, R.L. Suppression of corneal neovascularization with cyclosporine. Arch. Ophthalmol. 1992, 110, 405–407. [Google Scholar] [CrossRef] [PubMed]
  404. Benelli, U.; Ross, J.R.; Nardi, M.; Klintworth, G.K. Corneal neovascularization induced by xenografts or chemical cautery. Inhibition by cyclosporin A. Investig. Ophthalmol. Vis. Sci. 1997, 38, 274–282. [Google Scholar] [PubMed]
  405. Bucak, Y.Y.; Erdurmus, M.; Terzi, E.H.; Kükner, A.; Çelebi, S. Inhibitory effects of topical cyclosporine A 0.05% on immune-mediated corneal neovascularization in rabbits. Graefes. Arch. Clin. Exp. Ophthalmol. 2013, 251, 2555–2561. [Google Scholar] [CrossRef] [PubMed]
  406. Ulusoy, D.M.; Kahraman, N.; Balcıoğlu, E.; Duru, Z. Comparison of the inhibitory effect of topical cyclosporine A 0.1% and topical anti-VEGF application in an experimental model of corneal neovascularization. Arq. Bras. Oftalmol. 2022, 85, 19–24. [Google Scholar] [CrossRef] [PubMed]
  407. Cejkova, J.; Cejka, C.; Trosan, P.; Zajicova, A.; Sykova, E.; Holan, V. Treatment of alkali-injured cornea by cyclosporine A-loaded electrospun nanofibers—An alternative mode of therapy. Exp. Eye Res. 2016, 147, 128–137. [Google Scholar] [CrossRef] [PubMed]
  408. Perry, H.D.; Donnenfeld, E.D.; Kanellopoulos, A.J.; Grossman, G.A. Topical cyclosporin A in the management of postkeratoplasty glaucoma. Cornea 1997, 16, 284–288. [Google Scholar] [CrossRef] [PubMed]
  409. Perry, H.D.; Donnenfeld, E.D.; Acheampong, A.; Kanellopoulos, A.J.; Sforza, P.D.; D’Aversa, G.; Wallerstein, A.; Stern, M. Topical cyclosporine A in the management of postkeratoplasty glaucoma and corticosteroid-induced ocular hypertension and the penetration of topical 0.5% cyclosporine A into the cornea and anterior chamber. CLAO J. 1998, 24, 159–165. [Google Scholar] [PubMed]
  410. Okumuş, S.; Coşkun, E.; Tatar, M.G.; Kaydu, E.; Yayuspayi, R.; Comez, A.; Erbagci, I.; Gurler, B. Cyclosporine A 0.05% eye drops for the treatment of subepithelial infiltrates after epidemic keratoconjunctivitis. BMC Ophthalmol. 2012, 12, 42. [Google Scholar] [CrossRef] [PubMed]
  411. Steinmann, B.; Bruckner, P.; Superti-Furga, A. Cyclosporin A slows collagen triple-helix formation in vivo: Indirect evidence for a physiologic role of peptidyl-prolyl cis-trans-isomerase. J. Biol. Chem. 1991, 266, 1299–1303. [Google Scholar] [CrossRef] [PubMed]
  412. Fishel, R.; Barbul, A.; Wasserkrug, H.L.; Penberthy, L.T.; Rettura, G.; Efron, G. Cyclosporine A impairs wound healing in rats. J. Surg. Res. 1983, 34, 572–575. [Google Scholar] [CrossRef] [PubMed]
  413. Nemlander, A.; Ahonen, J.; Wiktorowicz, K.; von Willebrand, E.; Hekali, R.; Lalla, M.; Häyry, P. Effect of cyclosporine on wound healing. Transplantation 1983, 36, 1–5. [Google Scholar] [CrossRef] [PubMed]
  414. Recker, F.; Marquardt, K.; Redha, F.; Uhlschmid, G.; Largiadèr, F. Cyclosporine A impairs wound healing of ureterocystoneostomy in rats: Scanning electron microscopic examination. Urol. Res. 1989, 17, 21–26. [Google Scholar] [CrossRef] [PubMed]
  415. Petri, J.B.; Schurk, S.; Gebauer, S.; Haustein, U.F. Cyclosporine A delays wound healing and apoptosis and suppresses activin beta-A expression in rats. Eur. J. Dermatol. 1998, 8, 104–113. [Google Scholar] [PubMed]
  416. Aktaş, R.G.; Aktaş, S.; Yazgan, O.; Altaner, S. The effects of long-term low-dose cyclosporin A treatment on muscles and tendons: An experimental study. Ulus. Travma. Acil. Cerrahi. Derg. 2009, 15, 317–323. [Google Scholar] [PubMed]
  417. Lee, S.H.; Leem, H.S.; Jeong, S.M.; Lee, K. Bevacizumab accelerates corneal wound healing by inhibiting TGF-β2 expression in alkali-burned mouse cornea. BMB Rep. 2009, 42, 800–805. [Google Scholar] [CrossRef] [PubMed]
  418. Lee, K.J.; Lee, J.Y.; Lee, S.H.; Choi, T.H. Accelerating repaired basement membrane after bevacizumab treatment on alkali-burned mouse cornea. BMB Rep. 2013, 46, 195–200. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  419. Kim, T.I.; Chung, J.L.; Hong, J.P.; Min, K.; Seo, K.Y.; Kim, E.K. Bevacizumab application delays epithelial healing in rabbit cornea. Investig. Ophthalmol. Vis. Sci. 2009, 50, 4653–4659. [Google Scholar] [CrossRef] [PubMed]
  420. Kim, E.C.; Ryu, H.W.; Lee, H.J.; Kim, M.S. Bevacizumab eye drops delay corneal epithelial wound healing and increase the stromal response to epithelial injury in rats. Clin. Exp. Ophthalmol. 2013, 41, 694–701. [Google Scholar] [CrossRef] [PubMed]
  421. Al-Debasi, T.; Al-Bekairy, A.; Al-Katheri, A.; Al Harbi, S.; Mansour, M. Topical versus subconjunctival anti-vascular endothelial growth factor therapy (bevacizumab, ranibizumab and aflibercept) for treatment of corneal neovascularization. Saudi J. Ophthalmol. 2017, 31, 99–105. [Google Scholar] [CrossRef] [PubMed]
  422. Lai, S.C.; Loh, E.W.; Chiou, D.I.; Hong, C.T. Efficacy and safety of anti-vascular endothelial growth factor agents on corneal neovascularization: A meta-analysis. World J. Clin. Cases 2023, 11, 7337–7349. [Google Scholar] [CrossRef] [PubMed]
  423. Stevenson, W.; Cheng, S.F.; Dastjerdi, M.H.; Ferrari, G.; Dana, R. Corneal neovascularization and the utility of topical VEGF inhibition: Ranibizumab (Lucentis) vs bevacizumab (Avastin). Ocul. Surf. 2012, 10, 67–83. [Google Scholar] [CrossRef] [PubMed]
  424. Chang, J.H.; Garg, N.K.; Lunde, E.; Han, K.Y.; Jain, S.; Azar, D.T. Corneal neovascularization: An anti-VEGF therapy review. Surv. Ophthalmol. 2012, 57, 415–429. [Google Scholar] [CrossRef] [PubMed]
  425. Dastjerdi, M.H.; Al-Arfaj, K.; Nallasamy, N.; Hamrah, P.; Jurkunas, U.V.; Pineda, R., II; Pavan-Langston, D.; Dana, R. Topical bevacizumab in the treatment of corneal neovascularization: Results of a prospective, open-label, noncomparative study. Arch. Ophthalmol. 2009, 127, 381–389. [Google Scholar] [CrossRef] [PubMed]
  426. Kim, S.W.; Ha, B.J.; Kim, E.K.; Tchah, H.; Kim, T.I. The effect of topical bevacizumab on corneal neovascularization. Ophthalmology 2008, 115, e33–e38. [Google Scholar] [CrossRef] [PubMed]
  427. Bahar, I.; Kaiserman, I.; McAllum, P.; Rootman, D.; Slomovic, A. Subconjunctival bevacizumab injection for corneal neovascularization. Cornea 2008, 27, 142–147. [Google Scholar] [CrossRef] [PubMed]
  428. Chen, W.L.; Lin, C.T.; Lin, N.T.; Tu, I.H.; Li, J.W.; Chow, L.P.; Liu, K.R.; Hu, F.R. Subconjunctival injection of bevacizumab (Avastin) on corneal neovascularization in different rabbit models of corneal angiogenesis. Investig. Ophthalmol. Vis. Sci. 2009, 50, 1659–1665. [Google Scholar] [CrossRef] [PubMed]
  429. Park, Y.R.; Chung, S.K. Inhibitory effect of topical aflibercept on corneal neovascularization in rabbits. Cornea 2015, 34, 1303–1307. [Google Scholar] [CrossRef] [PubMed]
  430. Yoon, C.H.; Ko, J.H.; Lee, H.J.; Song, H.B.; Oh, J.Y. Subconjunctival aflibercept inhibits corneal angiogenesis and VEGFR-3+CD11b+ cells. Graefes. Arch. Clin. Exp. Ophthalmol. 2024, 262, 3881–3888. [Google Scholar] [CrossRef] [PubMed]
  431. Ferrari, G.; Dastjerdi, M.H.; Okanobo, A.; Cheng, S.F.; Amparo, F.; Nallasamy, N.; Dana, R. Topical ranibizumab as a treatment of corneal neovascularization. Cornea 2013, 32, 992–997. [Google Scholar] [CrossRef] [PubMed]
  432. Bucher, F.; Parthasarathy, A.; Bergua, A.; Onderka, J.; Regenfuss, B.; Cursiefen, C.; Bock, F. Topical ranibizumab inhibits inflammatory corneal hem- and lymphangiogenesis. Acta Ophthalmol. 2014, 92, 143–148. [Google Scholar] [CrossRef] [PubMed]
  433. Good, T.J.; Kimura, A.E.; Mandava, N.; Kahook, M.Y. Sustained elevation of intraocular pressure after intravitreal injections of anti-VEGF agents. Br. J. Ophthalmol. 2011, 95, 1111–1114. [Google Scholar] [CrossRef] [PubMed]
  434. Choi, D.Y.; Ortube, M.C.; McCannel, C.A.; Sarraf, D.; Hubschman, J.P.; McCannel, T.A.; Gorin, M.B. Sustained elevated intraocular pressures after intravitreal injection of bevacizumab, ranibizumab, and pegaptanib. Retina 2011, 31, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  435. Bakri, S.J.; McCannel, C.A.; Edwards, A.O.; Moshfeghi, D.M. Persistent ocular hypertension following intravitreal ranibizumab. Graefes. Arch. Clin. Exp. Ophthalmol. 2008, 246, 955–958. [Google Scholar] [CrossRef] [PubMed]
  436. Adelman, R.A.; Zheng, Q.; Mayer, H.R. Persistent ocular hypertension following intravitreal bevacizumab and ranibizumab injections. J. Ocul. Pharmacol. Ther. 2010, 26, 105–110. [Google Scholar] [CrossRef] [PubMed]
  437. Tseng, J.J.; Vance, S.K.; Della Torre, K.E.; Mendonca, L.S.; Cooney, M.J.; Klancnik, J.M.; Sorenson, J.A.; Freund, K.B. Sustained increased intraocular pressure related to intravitreal anti-vascular endothelial growth factor therapy for neovascular age-related macular degeneration. J. Glaucoma. 2012, 21, 241–247. [Google Scholar] [CrossRef] [PubMed]
  438. Mathalone, N.; Arodi-Golan, A.; Sar, S.; Wolfson, Y.; Shalem, M.; Lavi, I.; Geyer, O. Sustained elevation of intraocular pressure after intravitreal injections of bevacizumab in eyes with neovascular age-related macular degeneration. Graefes. Arch. Clin. Exp. Ophthalmol. 2012, 250, 1435–1440. [Google Scholar] [CrossRef] [PubMed]
  439. Hoang, Q.V.; Tsuang, A.J.; Gelman, R.; Mendonca, L.S.; Della Torre, K.E.; Jung, J.J.; Freund, K.B. Clinical predictors of sustained intraocular pressure elevation due to intravitreal anti-vascular endothelial growth factor therapy. Retina 2013, 33, 179–187. [Google Scholar] [CrossRef] [PubMed]
  440. Sniegowski, M.; Mandava, N.; Kahook, M.Y. Sustained intraocular pressure elevation after intravitreal injection of bevacizumab and ranibizumab associated with trabeculitis. Open Ophthalmol. J. 2010, 4, 28–29. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  441. Sahin, H.; Tholema, N.; Petersen, W.; Raschke, M.J.; Stange, R. Impaired biomechanical properties correlate with neoangiogenesis as well as VEGF and MMP-3 expression during rat patellar tendon healing. J. Orthop. Res. 2012, 30, 1952–1957. [Google Scholar] [CrossRef] [PubMed]
  442. Tempfer, H.; Kaser-Eichberger, A.; Lehner, C.; Gehwolf, R.; Korntner, S.; Kunkel, N.; Wagner, A.; Gruetz, M.; Heindl, L.M.; Schroedl, F.; et al. Bevacizumab improves Achilles tendon repair in a rat model. Cell. Physiol. Biochem. 2018, 46, 1148–1158. [Google Scholar] [CrossRef] [PubMed]
  443. Kusaba, Y.; Kumagai, K.; Ishikawa, K.; Choe, H.; Ike, H.; Kobayashi, N.; Inaba, Y. Bevacizumab promotes tenogenic differentiation and maturation of rat tendon-derived cells in vitro. PLoS ONE 2023, 18, e0293463. [Google Scholar] [CrossRef] [PubMed]
  444. Dallaudière, B.; Lempicki, M.; Pesquer, L.; Louedec, L.; Preux, P.M.; Meyer, P.; Hess, A.; Moreau-Durieux, M.H.; Hummel, V.; Larbi, A.; et al. Acceleration of tendon healing using US-guided intratendinous injection of bevacizumab: First pre-clinical study on a murine model. Eur. J. Radiol. 2013, 82, e823–e828. [Google Scholar] [CrossRef] [PubMed]
  445. Riggin, C.N.; Rodriguez, A.B.; Weiss, S.N.; Raja, H.A.; Chen, M.; Schultz, S.M.; Sehgal, C.M.; Soslowsky, L.J. Modulation of vascular response after injury in the rat Achilles tendon alters healing capacity. J. Orthop. Res. 2021, 39, 2000–2016. [Google Scholar] [CrossRef] [PubMed]
  446. Sikiric, P.; Seiwerth, S.; Rucman, R.; Turkovic, B.; Rokotov, D.S.; Brcic, L.; Sever, M.; Klicek, R.; Radic, B.; Drmic, D.; et al. Toxicity by NSAIDs. Counteraction by stable gastric pentadecapeptide BPC 157. Curr. Pharm. Des. 2013, 19, 76–83. [Google Scholar] [CrossRef] [PubMed]
  447. Folkman, J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. Med. 1995, 1, 27–31. [Google Scholar] [CrossRef] [PubMed]
  448. Klagsbrun, M.; Moses, M.A.M. Judah Folkman (1933–2008). Nature 2008, 451, 781. [Google Scholar] [CrossRef] [PubMed]
  449. Brower, V. Judah Folkman leaves expanding legacy. J. Natl. Cancer Inst. 2008, 100, 380–381. [Google Scholar] [CrossRef] [PubMed]
  450. Hanahan, D.; Weinberg, R.A. Retrospective: Judah Folkman (1933–2008). Science 2008, 319, 1055. [Google Scholar] [CrossRef] [PubMed]
  451. Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef] [PubMed]
  452. Tsujinaka, A.; Takai, Y.; Inoue, Y.; Tanito, M. A case of bilateral deep stromal corneal opacity and vascularization after use of multiple antiglaucoma medications including brimonidine tartrate ophthalmic solution. Acta Ophthalmol. 2019, 97, e948–e949. [Google Scholar] [CrossRef] [PubMed]
  453. Lusthaus, J.A.; Goldberg, I. Brimonidine and brinzolamide for treating glaucoma and ocular hypertension: A safety evaluation. Expert Opin. Drug Saf. 2017, 16, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
  454. Moshirfar, M.; Ziari, M.; Payne, C.J.; Stapley, S.R.; Ply, B.K.; Ronquillo, Y.C.; Hoopes, P.C. Bilateral lipid keratopathy in the setting of brimonidine tartrate use. Case Rep. Ophthalmol. Med. 2023, 2023, 8115622. [Google Scholar] [CrossRef] [PubMed]
  455. Purgert, R.J.; Meghpara, B.; Kolomeyer, N.N. Corneal subepithelial infiltrates associated with brimonidine use. Can. J. Ophthalmol. 2020, 55, e172–e174. [Google Scholar] [CrossRef] [PubMed]
  456. Chang, H.-L.; Kuo, B.-I.; Wu, J.-H.; Huang, W.-L.; Su, C.-C.; Chen, W.-L. Anti-glaucoma agents-induced pseudodendritic keratitis presumed to be herpetic simplex keratitis: A clinical case series. Sci. Rep. 2021, 11, 21443. [Google Scholar] [CrossRef] [PubMed]
  457. Hashimoto, Y.; Yokoo, S.; Usui, T.; Tsubota, Y.; Yamagami, S. High permeability and intercellular space widening with brimonidine tartrate eye drops in cultured stratified human corneal epithelial sheets. Cornea 2018, 37, 242–247. [Google Scholar] [CrossRef] [PubMed]
  458. Al Asmi, M.; Al Busaidi, A. Nummular keratitis: A rare manifestation of brimonidine allergy. Oman J. Ophthalmol. 2023, 17, 113–116. [Google Scholar] [CrossRef] [PubMed]
  459. Derick, R.J.; Robin, A.L.; Walters, T.R.; Barnebey, H.S.; Choplin, N.; Schuman, J.; Kelley, E.P.; Chen, K.; Stoecker, J.F. Brimonidine tartrate: A one-month dose response study. Ophthalmology 1997, 104, 131–136. [Google Scholar] [CrossRef] [PubMed]
  460. Schuman, J.S.; Horwitz, B.; Choplin, N.T.; David, R.; Albracht, D.; Chen, K.; Chronic Brimonidine Study Group. A 1-year study of brimonidine twice daily in glaucoma and ocular hypertension: A controlled, randomized, multicenter clinical trial. Arch. Ophthalmol. 1997, 115, 847–852. [Google Scholar] [CrossRef] [PubMed]
  461. Walters, T.R. Development and use of brimonidine in treating acute and chronic elevations of intraocular pressure: A review of safety, efficacy, dose response, and dosing studies. Surv. Ophthalmol. 1996, 41 (Suppl. S1), S19–S26. [Google Scholar] [CrossRef] [PubMed]
  462. Cantor, L.B. Brimonidine in the treatment of glaucoma and ocular hypertension. Ther. Clin. Risk Manag. 2006, 2, 337–346. [Google Scholar] [CrossRef] [PubMed]
  463. Stewart, W.C.; Ritch, R.; Shin, D.H.; Lehmann, R.P.; Shrader, C.E.; van Buskirk, E.M. Apraclonidine Adjunctive Therapy Study Group. The efficacy of apraclonidine as an adjunct to timolol therapy. Arch. Ophthalmol. 1995, 113, 287–292. [Google Scholar] [CrossRef] [PubMed]
  464. Stewart, W.C.; Laibovitz, R.; Horwitz, B.; Stewart, R.H.; Ritch, R.; Kottler, M. Apraclonidine Primary Therapy Study Group. A 90-day study of the efficacy and side effects of 0.25% and 0.5% apraclonidine vs. 0.5% timolol. Arch. Ophthalmol. 1996, 114, 938–942. [Google Scholar] [CrossRef] [PubMed]
  465. Stamer, W.D.; Huang, Y.; Seftor, R.E.; Svensson, S.S.; Snyder, R.W.; Regan, J.W. Cultured human trabecular meshwork cells express functional alpha 2A adrenergic receptors. Investig. Ophthalmol. Vis. Sci. 1996, 37, 2426–2433. [Google Scholar] [PubMed]
  466. Ooi, Y.H.; Oh, D.J.; Rhee, D.J. Analysis of alpha2-adrenergic receptors and effect of brimonidine on matrix metalloproteinases and their inhibitors in human ciliary body. Investig. Ophthalmol. Vis. Sci. 2009, 50, 4237–4243. [Google Scholar] [CrossRef] [PubMed]
  467. Prokosch, V.; Panagis, L.; Volk, G.F.; Dermon, C.; Thanos, S. Alpha2-adrenergic receptors and their core involvement in the process of axonal growth in retinal explants. Investig. Ophthalmol. Vis. Sci. 2010, 51, 6688–6699. [Google Scholar] [CrossRef] [PubMed]
  468. Reitsamer, H.A.; Posey, M.; Kiel, J.W. Effects of a topical alpha2 adrenergic agonist on ciliary blood flow and aqueous production in rabbits. Exp. Eye Res. 2006, 82, 405–415. [Google Scholar] [CrossRef] [PubMed]
  469. Danielson, P.; Alfredson, H.; Forsgren, S. In situ hybridization studies confirming recent findings of the existence of a local nonneuronal catecholamine production in human patellar tendinosis. Microsc. Res. Tech. 2007, 70, 908–911. [Google Scholar] [CrossRef]
  470. Romana-Souza, B.; Nascimento, A.P.; Brum, P.C.; Monte-Alto-Costa, A. Deletion of the α2A/α2C-adrenoceptors accelerates cutaneous wound healing in mice. Int. J. Exp. Pathol. 2014, 95, 330–341. [Google Scholar] [CrossRef] [PubMed]
  471. Backman, L.J.; Andersson, G.; Fong, G.; Alfredson, H.; Scott, A.; Danielson, P. Alpha-2 adrenergic stimulation triggers Achilles tenocyte hypercellularity: Comparison between two model systems. Scand. J. Med. Sci. Sports 2013, 23, 687–696. [Google Scholar] [CrossRef] [PubMed]
  472. Danielson, P.; Alfredson, H.; Forsgren, S. Studies on the importance of sympathetic innervation, adrenergic receptors, and a possible local catecholamine production in the development of patellar tendinopathy (tendinosis) in man. Microsc. Res. Tech. 2007, 70, 310–324. [Google Scholar] [CrossRef] [PubMed]
  473. Bjur, D.; Danielson, P.; Alfredson, H.; Forsgren, S. Immunohistochemical and in situ hybridization observations favor a local catecholamine production in the human Achilles tendon. Histol. Histopathol. 2008, 23, 197–208. [Google Scholar] [CrossRef] [PubMed]
  474. Jewson, J.L.; Lambert, G.W.; Storr, M.; Gaida, J.E. The sympathetic nervous system and tendinopathy: A systematic review. Sports Med. 2015, 45, 727–743. [Google Scholar] [CrossRef] [PubMed]
  475. Reidy, J.J.; Zarzour, J.; Thompson, H.W.; Beuerman, R.W. Effect of topical beta blockers on corneal epithelial wound healing in the rabbit. Br. J. Ophthalmol. 1994, 78, 377–380. [Google Scholar] [CrossRef] [PubMed]
  476. Pullar, C.E.; Zhao, M.; Song, B.; Pu, J.; Reid, B.; Ghoghawala, S.; McCaig, C.; Isseroff, R.R. Beta-adrenergic receptor agonists delay while antagonists accelerate epithelial wound healing: Evidence of an endogenous adrenergic network within the corneal epithelium. J. Cell. Physiol. 2007, 211, 261–272. [Google Scholar] [CrossRef] [PubMed]
  477. Ghoghawala, S.Y.; Mannis, M.J.; Pullar, C.E.; Rosenblatt, M.I.; Isseroff, R.R. Beta2-adrenergic receptor signaling mediates corneal epithelial wound repair. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1857–1863. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  478. Kasiri, A.; Ghomi, M.R.; Feghhi, M.; Farrahi, F.; Mirdehghan, M.S.; Hedayati, H. Topical Timolol Inhibits Corneal Neovascularization in Rabbits. Med. Hypothesis Discov. Innov. Ophthalmol. 2017, 6, 39–43. [Google Scholar] [PubMed] [PubMed Central]
  479. Kasiri, A.; Mirdehghan, M.S.; Farrahi, F.; Ostadian, F.; Feghhi, M.; Ghomi, M.R.; Jafari, A.M.; Rad, A.M.; Kasiri, N. Prevention of Corneal Neovascularization: A Preliminary Experimental Study in Rabbits. Med. Hypothesis Discov. Innov. Ophthalmol. 2020, 9, 47–55. [Google Scholar] [PubMed] [PubMed Central]
  480. Filippi, L.; de Libero, C.; Zamma Gallarati, B.; Fortunato, P.; Piozzi, E. Propranolol eye drops in patients with corneal neovascularization. Medicine 2018, 97, e13002. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  481. Ornek, F.; Turgut, B.; Doganay, S.; Daglioglu, M.C.; Demir, T.; Celiker, U.; Oner, A.O. Topical propranolol inhibits corneal neovascularization in experimental model of alkali burn in rats. Curr. Eye Res. 2011, 36, 312–318. [Google Scholar] [CrossRef] [PubMed]
  482. Turgut, B.; Ornek, F.; Doganay, S.; Daglioglu, M.C.; Demir, T.; Celiker, U.; Oner, A.O. Topical timolol inhibits corneal neovascularization in experimental model of alkali burn in rats. Curr. Eye Res. 2011, 36, 942–949. [Google Scholar] [CrossRef] [PubMed]
  483. Katz, I.M.; Hubbard, W.A.; Getson, A.J.; Gould, A.L. Intraocular pressure decrease in normal volunteers following timolol ophthalmic solution. Investig. Ophthalmol. 1976, 15, 489–492. [Google Scholar] [PubMed]
  484. Zimmerman, T.J.; Kaufman, H.E. Timolol. A β-adrenergic blocking agent for the treatment of glaucoma. Arch. Ophthalmol. 1977, 95, 601–604. [Google Scholar] [CrossRef] [PubMed]
  485. Zimmerman, T.J.; Kaufman, H.E. Timolol—Dose response and duration of action. Arch. Ophthalmol. 1977, 95, 605–607. [Google Scholar] [CrossRef] [PubMed]
  486. Dunham, C.N.; Spaide, R.F.; Dunham, G. The contralateral reduction of intraocular pressure by timolol. Br. J. Ophthalmol. 1994, 78, 38–40. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  487. Lee, S.; Remark, L.H.; Buchalter, D.B.; Josephson, A.M.; Wong, M.Z.; Litwa, H.P.; Ihejirika, R.; Leclerc, K.; Markus, D.; Yim, N.L.; et al. Propranolol reverses impaired fracture healing response observed with selective serotonin reuptake inhibitor treatment. J. Bone Miner. Res. 2020, 35, 932–941. [Google Scholar] [CrossRef] [PubMed]
  488. Mohammadi, A.A.; Bakhshaeekia, A.; Alibeigi, P.; Hasheminasab, M.J.; Tolide-ei, H.R.; Tavakkolian, A.R.; Mohammadi, M.K. Efficacy of propranolol in wound healing for hospitalized burn patients. J. Burn Care Res. 2009, 30, 1013–1017. [Google Scholar] [CrossRef] [PubMed]
  489. Romana-Souza, B.; Nascimento, A.P.; Monte-Alto-Costa, A. Low-dose propranolol improves cutaneous wound healing of burn-injured rats. Plast. Reconstr. Surg. 2008, 122, 1690–1699. [Google Scholar] [CrossRef] [PubMed]
  490. Al-Subaie, A.E.; Laurenti, M.; Abdallah, M.N.; Tamimi, I.; Yaghoubi, F.; Eimar, H.; Makhoul, N.; Tamimi, F. Propranolol enhances bone healing and implant osseointegration in rats’ tibiae. J. Clin. Periodontol. 2016, 43, 1160–1170. [Google Scholar] [CrossRef] [PubMed]
  491. Ali, A.; Herndon, D.N.; Mamachen, A.; Hasan, S.; Andersen, C.R.; Grogans, R.J.; Brewer, J.L.; Lee, J.O.; Heffernan, J.; Suman, O.E.; et al. Propranolol attenuates hemorrhage and accelerates wound healing in severely burned adults. Crit. Care 2015, 19, 217. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  492. Cahn, B.A.; Kaur, R.; Hirt, P.; Tchanque-Fossuo, C.; Dahle, S.E.; Kirsner, R.S.; Isseroff, R.R.; Lev-Tov, H. Use of topical timolol maleate as re-epithelialization agent for treatment of recalcitrant wounds of varying etiologies. J. Drugs Dermatol. 2020, 19, 1252–1256. [Google Scholar] [CrossRef] [PubMed]
  493. Dabiri, G.; Tiger, J.; Goreshi, R.; Fischer, A.; Iwamoto, S. Topical timolol may improve overall scar cosmesis in acute surgical wounds. Cutis 2017, 100, E27–E28. [Google Scholar] [PubMed]
  494. Konowal, A.; Morrison, J.C.; Brown, S.V.L.; Cooke, D.L.; Maguire, L.J.; Verdier, D.V.; Fraunfelder, F.T.; Dennis, R.F.; Epstein, R.J. Irreversible corneal decompensation in patients treated with topical dorzolamide. Am. J. Ophthalmol. 1999, 127, 403–406. [Google Scholar] [CrossRef] [PubMed]
  495. Zhao, J.C.; Chen, T. Brinzolamide induced reversible corneal decompensation. Br. J. Ophthalmol. 2005, 89, 389–390. [Google Scholar] [CrossRef] [PubMed]
  496. Kwak, J.; Jun, J.H. Reversible corneal decompensation caused by a topical dorzolamide/timolol fixed combination after Descemet stripping automated endothelial keratoplasty. Cornea 2022, 41, 1171–1173. [Google Scholar] [CrossRef] [PubMed]
  497. Giasson, C.J.; Nguyen, T.Q.; Boisjoly, H.M.; Lesk, M.R.; Amyot, M.; Charest, M. Dorzolamide and corneal recovery from edema in patients with glaucoma or ocular hypertension. Am. J. Ophthalmol. 2000, 129, 144–150. [Google Scholar] [CrossRef] [PubMed]
  498. Wirtitsch, M.G.; Findl, O.; Kiss, B.; Petternel, V.; Heinzl, H.; Drexler, W. Short-term effect of dorzolamide hydrochloride on central corneal thickness in humans with cornea guttata. Arch. Ophthalmol. 2003, 121, 621–625. [Google Scholar] [CrossRef] [PubMed][Green Version]
  499. Agrawal, V. First aid for complications of infectious keratitis. Indian J. Ophthalmol. 2008, 56, 221–222. [Google Scholar] [CrossRef] [PubMed]
  500. Manabe, Y.; Sawada, A.; Mochizuki, K. Corneal sterile infiltration induced by topical use of ocular hypotensive agent. Eur. J. Ophthalmol. 2020, 30, NP23–NP25. [Google Scholar] [CrossRef] [PubMed]
  501. Kasuya, Y.; Sano, I.; Makino, S.; Kawashima, H. Corneal opacity induced by antiglaucoma agents other than brimonidine tartrate. Case Rep. Ophthalmol. Med. 2020, 2020, 4803651. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  502. Maruyama, Y.; Ikeda, Y.; Yokoi, N.; Mori, K.; Kato, H.; Ueno, M.; Kinoshita, S.; Sotozono, C. Severe corneal disorders developed after brimonidine tartrate ophthalmic solution use. Cornea 2017, 36, 1567–1569. [Google Scholar] [CrossRef] [PubMed]
  503. Balfour, J.A.; Wilde, M.I. Dorzolamide. A review of its pharmacology and therapeutic potential in the management of glaucoma and ocular hypertension. Drugs Aging 1997, 10, 384–403. [Google Scholar] [CrossRef] [PubMed]
  504. Barker, H.L.; Aaltonen, M.; Pan, P.; Vähätupa, M.; Kaipiainen, P.; May, U.; Prince, S.; Uusitalo-Järvinen, H.; Waheed, A.; Pastoreková, S.; et al. Role of carbonic anhydrases in skin wound healing. Exp. Mol. Med. 2017, 49, e334. [Google Scholar] [CrossRef] [PubMed]
  505. Gallorini, M.; Berardi, A.C.; Ricci, A.; Antonetti Lamorgese Passeri, C.; Zara, S.; Oliva, F.; Cataldi, A.; Carta, F.; Carradori, S. Dual Acting Carbon Monoxide Releasing Molecules and Carbonic Anhydrase Inhibitors Differentially Modulate Inflammation in Human Tenocytes. Biomedicines 2021, 9, 141. [Google Scholar] [CrossRef] [PubMed]
  506. Er, H. The effect of topical parasympathomimetics on corneal epithelial healing in rabbits. Doc. Ophthalmol. 1997, 93, 327–335. [Google Scholar] [CrossRef] [PubMed]
  507. Oztürk, F.; Kurt, E.; Inan, U.U.; Emiroğlu, L.; Ilker, S.S. The effects of acetylcholine and propolis extract on corneal epithelial wound healing in rats. Cornea 1999, 18, 466–471. [Google Scholar] [CrossRef] [PubMed]
  508. Robin, J.B.; Kash, R.L.; Azen, S.P.; Schanzlin, D.J. Lack of effect of pilocarpine on corneal epithelial wound healing. Curr. Eye Res. 1984, 3, 403–406. [Google Scholar] [CrossRef] [PubMed]
  509. Yuan, X.L.; Wen, Q.; Zhang, M.Y.; Fan, T.J. Cytotoxicity of pilocarpine to human corneal stromal cells and its underlying cytotoxic mechanisms. Int. J. Ophthalmol. 2016, 9, 505–511. [Google Scholar] [CrossRef]
  510. Coles, W.H. Pilocarpine toxicity. Effects on the rabbit corneal endothelium. Arch. Ophthalmol. 1975, 93, 36–41. [Google Scholar] [CrossRef] [PubMed]
  511. Liu, G.S.; Trope, G.E.; Basu, P.K. The toxic effects of pilocarpine gel and drops on rabbit cornea. Curr. Eye Res. 1989, 8, 637–648. [Google Scholar] [CrossRef] [PubMed]
  512. Jay, J.L.; MacDonald, M. Effects of intraocular miotics on cultured bovine corneal endothelium. Br. J. Ophthalmol. 1978, 62, 815–820. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  513. Massry, G.G.; Assil, K.K. Pilocarpine-associated allograft rejection in postkeratoplasty patients. Cornea 1995, 14, 202–205. [Google Scholar] [CrossRef] [PubMed]
  514. Li, S.; Ding, G.; Sun, Y.; Zhang, C. Impact of different oxygen supply methods on the healing of corneal epithelial wound and the level of acetylcholine. J. Ophthalmol. 2021, 2021, 4737479. [Google Scholar] [CrossRef] [PubMed]
  515. Seibold, L.K.; Wagner, B.D.; Lynch, A.M.; Kahook, M.Y. The diurnal and nocturnal effects of pilocarpine on intraocular pressure in patients receiving prostaglandin analog monotherapy. J. Ocul. Pharmacol. Ther. 2018, 34, 590–595. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  516. Kannarr, S.; El-Harazi, S.M.; Moshirfar, M.; Lievens, C.; Kim, J.L.; Peace, J.H.; Safyan, E.; Liu, H.; Zheng, S.; Robinson, M.R. Safety and efficacy of twice-daily pilocarpine HCl in presbyopia: The VIRGO Phase 3, randomized, double-masked, controlled study. Am. J. Ophthalmol. 2023, 253, 189–200. [Google Scholar] [CrossRef] [PubMed]
  517. Brown, S.V.; Thomas, J.V.; Belcher, C.D., 3rd; Simmons, R.J. Effect of pilocarpine in treatment of intraocular-pressure elevation following neodymium: YAG laser posterior capsulotomy. Am. J. Ophthalmol. 1985, 92, 354–359. [Google Scholar] [CrossRef] [PubMed]
  518. Zimmer, A.; Mutschler, E.; Lambrecht, G.; Mayer, D.; Kreuter, J. Pharmacokinetic and pharmacodynamic aspects of an ophthalmic pilocarpine nanoparticle-delivery-system. Pharm. Res. 1994, 11, 1435–1442. [Google Scholar] [CrossRef] [PubMed]
  519. Wang, Z.; Chen, Y.; Xiao, H.; Li, S.; Zhang, T.; Hu, J.; Lu, H.; Xie, H. The enhancement effect of acetylcholine and pyridostigmine on bone-tendon interface healing in a murine rotator cuff model. Am. J. Sports Med. 2021, 49, 909–917. [Google Scholar] [CrossRef] [PubMed]
  520. Fong, G.; Backman, L.J.; Andersson, G.; Scott, A.; Danielson, P. Human tenocytes are stimulated to proliferate by acetylcholine through an EGFR signalling pathway. Cell Tissue Res. 2013, 351, 465–475. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  521. Takada, Y.; Yasuda, S.; Sumioka, T.; Okada, Y.; Tamura, T.; Yamanaka, O.; Saika, S. Effects of Ripasudil Hydrochloride on Epithelial Repair in a Mouse Cornea. Curr. Eye Res. 2024, 49, 1215–1222. [Google Scholar] [CrossRef] [PubMed]
  522. Okumura, N.; Okazaki, Y.; Inoue, R.; Kakutani, K.; Nakano, S.; Kinoshita, S.; Koizumi, N. Effect of the Rho-Associated Kinase Inhibitor Eye Drop (Ripasudil) on Corneal Endothelial Wound Healing. Investig. Ophthalmol. Vis. Sci. 2016, 57, 1284–1292. [Google Scholar] [CrossRef] [PubMed]
  523. Bronner, A.W. Case Report: Topical Netarsudil in the Treatment of a Neurotrophic Corneal Ulcer. Optom. Vis. Sci. 2022, 99, 589–592. [Google Scholar] [CrossRef] [PubMed]
  524. Chen, S.L.; LoBue, S.A.; Goyal, H. Case Report: The Use of Netarsudil to Improve Corneal Edema after Laser Peripheral Iridotomy and Descemet’s Membrane Endothelial Keratoplasty. Am. J. Ophthalmol. Case Rep. 2021, 22, 101087. [Google Scholar] [CrossRef] [PubMed]
  525. Inomata, T.; Fujimoto, K.; Okumura, Y.; Zhu, J.; Fujio, K.; Shokirova, H.; Miura, M.; Okano, M.; Funaki, T.; Sung, J.; et al. Novel Immunotherapeutic Effects of Topically Administered Ripasudil (K-115) on Corneal Allograft Survival. Sci. Rep. 2020, 10, 19817. [Google Scholar] [CrossRef] [PubMed]
  526. Zeng, P.; Pi, R.B.; Li, P.; Chen, R.X.; Lin, L.M.; He, H.; Zhou, S.Y. Fasudil Hydrochloride, a Potent ROCK Inhibitor, Inhibits Corneal Neovascularization after Alkali Burns in Mice. Mol. Vis. 2015, 21, 688–698. [Google Scholar] [PubMed] [PubMed Central]
  527. Sood, T.; Chauhan, N.; Shree, A. Corneal Vascularisation and Haemorrhage Associated with Netarsudil Drops. Clin. Exp. Optom. 2025, 108, 643–644. [Google Scholar] [CrossRef] [PubMed]
  528. Kusuhara, S.; Nakamura, M. Ripasudil Hydrochloride Hydrate in the Treatment of Glaucoma: Safety, Efficacy, and Patient Selection. Clin. Ophthalmol. 2020, 14, 1229–1236. [Google Scholar] [CrossRef] [PubMed]
  529. Kiderlen, S.; Polzer, C.; Rädler, J.O.; Docheva, D.; Clausen-Schaumann, H.; Sudhop, S. Age Related Changes in Cell Stiffness of Tendon Stem/Progenitor Cells and a Rejuvenating Effect of ROCK-Inhibition. Biochem. Biophys. Res. Commun. 2019, 509, 839–844. [Google Scholar] [CrossRef] [PubMed]
  530. Melzer, M.; Schubert, S.; Müller, S.F.; Geyer, J.; Hagen, A.; Niebert, S.; Burk, J.R. Rho/ROCK Inhibition Promotes TGF-β3-Induced Tenogenic Differentiation in Mesenchymal Stromal Cells. Stem Cells Int. 2021, 2021, 8284690. [Google Scholar] [CrossRef] [PubMed]
  531. Liang, H.; Baudouin, C.; Daull, P.; Garrigue, J.-S.; Buggage, R.; Brignole-Baudouin, F. In Vitro and In Vivo Evaluation of a Preservative-Free Cationic Emulsion of Latanoprost in Corneal Wound Healing Models. Cornea 2012, 31, 1319–1329. [Google Scholar] [CrossRef] [PubMed]
  532. Daull, P.; Buggage, R.; Lambert, G.; Faure, M.-O.; Serle, J.; Wang, R.-F.; Garrigue, J.-S. A Comparative Study of a Preservative-Free Latanoprost Cationic Emulsion (Catioprost) and a BAK-Preserved Latanoprost Solution in Animal Models. J. Ocul. Pharmacol. Ther. 2012, 28, 515–523. [Google Scholar] [CrossRef] [PubMed]
  533. Pauly, A.; Roubeix, C.; Liang, H.; Brignole-Baudouin, F.; Baudouin, C. In Vitro and In Vivo Comparative Toxicological Study of a New Preservative-Free Latanoprost Formulation. Investig. Ophthalmol. Vis. Sci. 2012, 53, 8172–8180. [Google Scholar] [CrossRef] [PubMed]
  534. Daull, P.; Feraille, L.; Elena, P.-P.; Garrigue, J.-S. Comparison of the Anti-Inflammatory Effects of Artificial Tears in a Rat Model of Corneal Scraping. J. Ocul. Pharmacol. Ther. 2016, 32, 109–118. [Google Scholar] [CrossRef] [PubMed]
  535. Nagai, N.; Murao, T.; Okamoto, N.; Ito, Y. Comparison of Corneal Wound Healing Rates after Instillation of Commercially Available Latanoprost and Travoprost in Rat Debrided Corneal Epithelium. J. Oleo Sci. 2010, 59, 135–141. [Google Scholar] [CrossRef] [PubMed]
  536. Daull, P.; Garrigue, J.-S.; Liang, H.; Baudouin, C. Use of a Cationic Emulsion of Latanoprost to Treat Glaucoma Patients with Ocular Surface Disease: A Preclinical Review. J. Ocul. Pharmacol. Ther. 2023, 39, 240–251. [Google Scholar] [CrossRef] [PubMed]
  537. Dutca, L.M.; Rudd, D.; Robles, V.; Galor, A.; Garvin, M.K.; Anderson, M.G. Effects of Sustained Daily Latanoprost Application on Anterior Chamber Anatomy and Physiology in Mice. Sci. Rep. 2018, 8, 13088. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  538. Sudesh, S.; Cohen, E.J.; Rapuano, C.J.; Wilson, R.P. Corneal Toxicity Associated with Latanoprost. Arch. Ophthalmol. 1999, 117, 539–540. [Google Scholar] [CrossRef] [PubMed]
  539. Wand, M.; Gilbert, C.M.; Liesegang, T.J. Latanoprost and Herpes Simplex Keratitis. Am. J. Ophthalmol. 1999, 127, 602–604. [Google Scholar] [CrossRef] [PubMed]
  540. Kaufman, H.E.; Varnell, E.D.; Thompson, H.W.; Bazan, N.G. Latanoprost Increases the Severity and Recurrence of Herpetic Keratitis in the Rabbit. Am. J. Ophthalmol. 1999, 127, 531–536. [Google Scholar] [CrossRef] [PubMed]
  541. Orhan, M.; Kerimoğlu, H.; Irkeç, M. Corneal Neovascularization Possibly Associated with Latanoprost Therapy. Eur. J. Ophthalmol. 2003, 13, 88–90. [Google Scholar] [CrossRef] [PubMed]
  542. Gibran, S.K. Unilateral Drug-Induced Ocular Pseudopemphigoid. Eye 2004, 18, 1270. [Google Scholar] [CrossRef] [PubMed][Green Version]
  543. Watanabe, K.; Hayasaka, S.; Hayasaka, Y.; Nagaki, Y.; Watanabe, K. Cystoid macular edema associated with latanoprost use in a pseudophakic eye with a history of surgical complications. Jpn. J. Ophthalmol. 2003, 47, 110–112. [Google Scholar] [CrossRef] [PubMed]
  544. Chen, W.; Dong, N.; Huang, C.; Zhang, Z.; Hu, J.; Xie, H.; Pan, J.; Liu, Z. Corneal Alterations Induced by Topical Application of Commercial Latanoprost, Travoprost and Bimatoprost in Rabbit. PLoS ONE 2014, 9, e89205. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  545. Schwartz, S.; George, J.; Ben-Shoshan, J.; Luboshits, G.; Avni, I.; Levkovitch-Verbin, H.; Ziv, H.; Rosner, M. Drug Modification of Angiogenesis in a Rat Cornea Model. Investig. Ophthalmol. Vis. Sci. 2008, 49, 250–254. [Google Scholar] [CrossRef] [PubMed]
  546. Larsson, L.I. Intraocular Pressure over 24 Hours after Single-Dose Administration of Latanoprost 0.005% in Healthy Volunteers: A Randomized, Double-Masked, Placebo-Controlled, Cross-Over Single-Center Study. Acta Ophthalmol. Scand. 2001, 79, 567–571. [Google Scholar] [CrossRef] [PubMed]
  547. Ferry, S.T.; Weinhold, P.S.; Afshari, H.M.; Kerns, J.M.; Simpson, L.A.; Corona, B.T.; Vanderburgh, P.M.; Godges, J.J.; Wilkins, B.; Lieber, R.L.; et al. Effect of Prostaglandin E2 Injection on the Structural Properties of the Rat Patellar Tendon. J. Appl. Physiol. 2012, 113, 1613–1621. [Google Scholar] [CrossRef] [PubMed]
  548. Zhang, J.; Wang, J.H.-C. Prostaglandin E2 (PGE2) Exerts Biphasic Effects on Human Tendon Stem Cells. PLoS ONE 2014, 9, e87706. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  549. Sharif, N.A. Degeneration of retina-brain components and connections in glaucoma: Disease causation and treatment options for eyesight preservation. Curr. Res. Neurobiol. 2022, 3, 100037. [Google Scholar] [CrossRef] [PubMed]
  550. Nathan, J. Hippocrates to Duke-Elder: An overview of the history of glaucoma. Clin. Exp. Optom. 2000, 83, 116–118. [Google Scholar] [CrossRef] [PubMed][Green Version]
  551. Klimko, P.G.; Sharif, N.A. Discovery, characterization and clinical utility of prostaglandin agonists for the treatment of glaucoma. Br. J. Pharmacol. 2019, 176, 1051–1058. [Google Scholar] [CrossRef] [PubMed]
  552. Bartels, S.P.; Roth, H.O.; Jumblatt, M.M.; Neufeld, A.H. Pharmacological effects of topical timolol in the rabbit eye. Investig. Ophthalmol. Vis. Sci. 1980, 19, 1189–1197. [Google Scholar] [PubMed]
  553. Katz, I.M. Beta-blockers and the eye: An overview. Ann. Ophthalmol. 1978, 10, 847–850. [Google Scholar] [PubMed]
  554. Knezevic, M.; Gojkovic, S.; Krezic, I.; Zizek, H.; Malekinusic, D.; Vrdoljak, B.; Vranes, H.; Knezevic, T.; Barisic, I.; Horvat Pavlov, K.; et al. Occlusion of the superior mesenteric artery in rats reversed by collateral pathways activation: Gastric pentadecapeptide BPC 157 therapy counteracts multiple organ dysfunction syndrome; intracranial, portal and caval hypertension; and aortal hypotension. Biomedicines 2021, 9, 609. [Google Scholar] [CrossRef] [PubMed]
  555. Knezevic, M.; Gojkovic, S.; Krezic, I.; Zizek, H.; Malekinusic, D.; Vrdoljak, B.; Knezevic, T.; Vranes, H.; Drmic, D.; Staroveski, M.; et al. Occluded superior mesenteric artery and vein. Therapy with the stable gastric pentadecapeptide BPC 157. Biomedicines 2021, 9, 792. [Google Scholar] [CrossRef] [PubMed]
  556. Knezevic, M.; Gojkovic, S.; Krezic, I.; Zizek, H.; Vranes, H.; Malekinusic, D.; Vrdoljak, B.; Knezevic, T.; Pavlov, K.H.; Drmic, D.; et al. Complex syndrome of the complete occlusion of the end of the superior mesenteric vein, opposed with the stable gastric pentadecapeptide BPC 157 in rats. Biomedicines 2021, 9, 1029. [Google Scholar] [CrossRef] [PubMed]
  557. Kolovrat, M.; Gojkovic, S.; Krezic, I.; Malekinusic, D.; Vrdoljak, B.; Kasnik Kovac, K.; Kralj, T.; Drmic, D.; Barisic, I.; Horvat Pavlov, K.; et al. Pentadecapeptide BPC 157 resolves Pringle maneuver in rats, both ischemia and reperfusion. World J. Hepatol. 2020, 12, 184–206. [Google Scholar] [CrossRef] [PubMed]
  558. Gojkovic, S.; Krezic, I.; Vrdoljak, B.; Malekinusic, D.; Barisic, I.; Petrovic, A.; Horvat Pavlov, K.; Kolovrat, M.; Duzel, A.; Knezevic, M.; et al. Pentadecapeptide BPC 157 resolves suprahepatic occlusion of the inferior caval vein, Budd-Chiari syndrome model in rats. World J. Gastrointest. Pathophysiol. 2020, 11, 1–19. [Google Scholar] [CrossRef] [PubMed]
  559. Vukojevic, J.; Siroglavic, M.; Kasnik, K.; Kralj, T.; Stancic, D.; Kokot, A.; Kolaric, D.; Drmic, D.; Sever, A.Z.; Barisic, I.; et al. Rat inferior caval vein (ICV) ligature and particular new insights with the stable gastric pentadecapeptide BPC 157. Vasc. Pharmacol. 2018, 106, 54–66. [Google Scholar] [CrossRef] [PubMed]
  560. Gojkovic, S.; Krezic, I.; Vranes, H.; Zizek, H.; Drmic, D.; Pavlov, K.H.; Petrovic, A.; Batelja, L.; Milavic, M.; Sikiric, S.; et al. BPC 157 therapy and the permanent occlusion of the superior sagittal sinus in rat. Vascular recruitment. Biomedicines 2021, 9, 744. [Google Scholar] [CrossRef] [PubMed]
  561. Vukojevic, J.; Vrdoljak, B.; Malekinusic, D.; Siroglavic, M.; Milavic, M.; Kolenc, D.; Boban Blagaic, A.; Batelja, L.; Drmic, D.; Seiwerth, S.; et al. The effect of pentadecapeptide BPC 157 on hippocampal ischemia/reperfusion injuries in rats. Brain Behav. 2020, 10, e01726. [Google Scholar] [CrossRef] [PubMed]
  562. Tepes, M.; Gojkovic, S.; Krezic, I.; Zizek, H.; Vranes, H.; Madzar, Z.; Santak, G.; Batelja, L.; Milavic, M.; Sikiric, S.; et al. Stable gastric pentadecapeptide BPC 157 therapy for primary abdominal compartment syndrome. Front. Pharmacol. 2021, 12, 718147. [Google Scholar] [CrossRef] [PubMed]
  563. Tepes, M.; Krezic, I.; Vranes, H.; Smoday, I.M.; Kalogjera, L.; Zizek, H.; Vukovic, V.; Oroz, K.; Kovac, K.K.; Madzar, Z.; et al. Stable gastric pentadecapeptide BPC 157: Effect on reperfusion following maintained intra-abdominal hypertension (grade III and grade IV) in rats. Pharmaceuticals 2023, 16, 1554. [Google Scholar] [CrossRef] [PubMed]
  564. Smoday, I.M.; Petrovic, I.; Kalogjera, L.; Vranes, H.; Zizek, H.; Krezic, I.; Gojkovic, S.; Skorak, I.; Hriberski, K.; Brizic, I.; et al. Therapy effect of the stable gastric pentadecapeptide BPC 157 on acute pancreatitis as vascular failure-induced severe peripheral and central syndrome in rats. Biomedicines 2022, 10, 1299. [Google Scholar] [CrossRef] [PubMed]
  565. Kalogjera, L.; Krezic, I.; Smoday, I.M.; Vranes, H.; Zizek, H.; Yago, H.; Oroz, K.; Vukovic, V.; Kavelj, I.; Novosel, L.; et al. Stomach perforation-induced general occlusion/occlusion-like syndrome and stable gastric pentadecapeptide BPC 157 therapy effect. World J. Gastroenterol. 2023, 29, 4289–4316. [Google Scholar] [CrossRef] [PubMed]
  566. Strbe, S.; Gojkovic, S.; Krezic, I.; Zizek, H.; Vranes, H.; Barisic, I.; Strinic, D.; Orct, T.; Vukojevic, J.; Ilic, S.; et al. Over-dose lithium toxicity as an occlusive-like syndrome in rats and gastric pentadecapeptide BPC 157. Biomedicines 2021, 9, 1506. [Google Scholar] [CrossRef] [PubMed]
  567. Gojkovic, S.; Krezic, I.; Vranes, H.; Zizek, H.; Drmic, D.; Batelja Vuletic, L.; Milavic, M.; Sikiric, S.; Stilinovic, I.; Simeon, P.; et al. Robert’s intragastric alcohol-induced gastric lesion model as an escalated general peripheral and central syndrome, counteracted by the stable gastric pentadecapeptide BPC 157. Biomedicines 2021, 9, 1300. [Google Scholar] [CrossRef] [PubMed]
  568. Smoday, I.M.; Krezic, I.; Kalogjera, L.; Vukovic, V.; Zizek, H.; Skoro, M.; Kovac, K.K.; Vranes, H.; Barisic, I.; Sikiric, S.; et al. Pentadecapeptide BPC 157 as therapy for inferior caval vein embolization: Recovery of sodium laurate-post-embolization syndrome in rats. Pharmaceuticals 2023, 16, 1507. [Google Scholar] [CrossRef] [PubMed]
  569. Premuzic Mestrovic, I.; Smoday, I.M.; Kalogjera, L.; Krezic, I.; Zizek, H.; Vranes, H.; Vukovic, V.; Oroz, K.; Skorak, I.; Brizic, I.; et al. Antiarrhythmic sotalol, occlusion/occlusion-like syndrome in rats, and stable gastric pentadecapeptide BPC 157 therapy. Pharmaceuticals 2023, 16, 977. [Google Scholar] [CrossRef] [PubMed]
  570. Strbe, S.; Smoday, I.M.; Krezic, I.; Kalogjera, L.; Vukovic, V.; Zizek, H.; Gojkovic, S.; Vranes, H.; Barisic, I.; Sikiric, S.; et al. Innate vascular failure by application of neuroleptics, amphetamine, and domperidone rapidly induced severe occlusion/occlusion-like syndromes in rats and stable gastric pentadecapeptide BPC 157 as therapy. Pharmaceuticals 2023, 16, 788. [Google Scholar] [CrossRef] [PubMed]
  571. Barisic, I.; Balenovic, D.; Udovicic, M.; Bardak, D.; Strinic, D.; Vlainic, J.; Vranes, H.; Smoday, I.M.; Krezic, I.; Milavic, M.; et al. Stable gastric pentadecapeptide BPC 157 may counteract myocardial infarction induced by isoprenaline in rats. Biomedicines 2022, 10, 265. [Google Scholar] [CrossRef] [PubMed]
  572. Gamulin, O.; Oroz, K.; Coric, L.; Krajacic, M.; Skrabic, M.; Dretar, V.; Strbe, S.; Talapko, J.; Juzbasic, M.; Krezic, I.; et al. Fourier transform infrared spectroscopy reveals molecular changes in blood vessels of rats treated with pentadecapeptide BPC 157. Biomedicines 2022, 10, 3130. [Google Scholar] [CrossRef] [PubMed]
  573. Sugioka, K.; Fukuda, K.; Nishida, T.; Kusaka, S. The fibrinolytic system in the cornea: A key regulator of corneal wound healing and biological defense. Exp. Eye Res. 2021, 204, 108459. [Google Scholar] [CrossRef] [PubMed]
  574. Haney, A.F.; Doty, E. The formation of coalescing peritoneal adhesions requires injury to both contacting peritoneal surfaces. Fertil. Steril. 1994, 61, 767–775. [Google Scholar] [CrossRef] [PubMed]
  575. Cesar, L.B.; Gojkovic, S.; Krezic, I.; Malekinusic, D.; Zizek, H.; Vuletic, L.B.; Petrovic, A.; Pavlov, K.H.; Drmic, D.; Kokot, A.; et al. Bowel adhesion and therapy with the stable gastric pentadecapeptide BPC 157, L-NAME and L-arginine in rats. World J. Gastrointest. Pharmacol. Ther. 2020, 11, 93–109. [Google Scholar] [CrossRef] [PubMed]
  576. Davey, A.K.; Maher, P.J. Surgical adhesions: A timely update, a great challenge for the future. J. Minim. Invasive Gynecol. 2007, 14, 15–22. [Google Scholar] [CrossRef] [PubMed]
  577. Collen, D. On the regulation and control of fibrinolysis. Thromb. Haemost. 1980, 43, 77–89. [Google Scholar] [CrossRef] [PubMed]
  578. Raftery, A.T. Effect of peritoneal trauma on peritoneal fibrinolytic activity and intraperitoneal adhesion formation. Eur. Surg. Res. 1981, 13, 397–401. [Google Scholar] [CrossRef] [PubMed]
  579. Stupnisek, M.; Franjic, S.; Drmic, D.; Hrelec, M.; Kolenc, D.; Radic, B.; Bojic, D.; Vcev, A.; Seiwerth, S.; Sikiric, P. Pentadecapeptide BPC 157 reduces bleeding time and thrombocytopenia after amputation in rats treated with heparin, warfarin or aspirin. Thromb. Res. 2012, 129, 652–659. [Google Scholar] [CrossRef] [PubMed]
  580. Hrelec, M.; Klicek, R.; Brcic, L.; Brcic, I.; Cvjetko, I.; Seiwerth, S.; Sikiric, P. Abdominal aorta anastomosis in rats and stable gastric pentadecapeptide BPC 157, prophylaxis and therapy. J. Physiol. Pharmacol. 2009, 60 (Suppl. S7), 161–165. [Google Scholar] [PubMed]
  581. Konosic, S.; Petricevic, M.; Ivancan, V.; Konosic, L.; Goluza, E.; Krtalic, B.; Drmic, D.; Stupnisek, M.; Seiwerth, S.; Sikiric, P. Intragastric application of aspirin, clopidogrel, cilostazol, and BPC 157 in rats: Platelet aggregation and blood clot. Oxid. Med. Cell. Longev. 2019, 2019, 9084643. [Google Scholar] [CrossRef] [PubMed]
  582. Ilić, S.; Brčić, I.; Mester, M.; Filipović, M.; Sever, M.; Kliček, R.; Barišić, I.; Radić, B.; Zoričić, Z.; Bilić, V.; et al. Over-dose insulin and stable gastric pentadecapeptide BPC 157. Attenuated gastric ulcers, seizures, brain lesions, hepatomegaly, fatty liver, breakdown of liver glycogen, profound hypoglycemia and calcification in rats. J. Physiol. Pharmacol. 2009, 60 (Suppl. S7), 107–114. [Google Scholar] [PubMed]
  583. Xu, C.; Sun, L.; Ren, F.; Huang, P.; Tian, Z.; Cui, J.; Zhang, W.; Wang, S.; Zhang, K.; He, L.; et al. Preclinical safety evaluation of body protective compound-157, a potential drug for treating various wounds. Regul. Toxicol. Pharmacol. 2020, 114, 104665. [Google Scholar] [CrossRef] [PubMed]
  584. Lee, E.; Burgess, K. Safety of intravenous infusion of BPC157 in humans: A pilot study. Altern. Ther. Health Med. 2025, 31, 20–24. [Google Scholar] [PubMed]
Figure 1. Cytoprotective triad: corneal ulcer healing, corneal neovascularization (CNV) control, and intraocular pressure (IOP) regulation. Tendon healing (avascular parallel) is included as an external validation axis for avascular tissue-specific efficacy.
Figure 1. Cytoprotective triad: corneal ulcer healing, corneal neovascularization (CNV) control, and intraocular pressure (IOP) regulation. Tendon healing (avascular parallel) is included as an external validation axis for avascular tissue-specific efficacy.
Pharmaceuticals 18 01822 g001
Table 1. Preclinical evidence of BPC 157 in eye therapy.
Table 1. Preclinical evidence of BPC 157 in eye therapy.
Condition/ModelTherapeutic Effect of BPC 157Reference
Glaucoma (rats)Rapidly lowers elevated intraocular pressure to normal levels; preserves retinal structure[9,35]
Pupil function disturbances
(atropine-induced mydriasis,
L-NAME/L-arginine-induced miosis) (rats, guinea pigs)
Restores normal pupil function; effect
dependent on NO system interaction
[36]
Retinal ischemia (L-NAME retrobulbar application, rats)Recovers retinal function and integrity;
prevents ischemic damage
[37]
Corneal abrasion (complete
epithelial defect, rats)
Promotes rapid healing; maintains corneal transparency[38]
Corneal ulceration/perforating corneal
injury (rats)
Facilitates healing of perforated corneal injury; prevents ulcer progression; counteracts corneal neovascularization[3]
Dry eye (lacrimal gland
removal, rats)
Counteracts tear deficiency and associated damage[39]
Corneal insensitivity (local
anesthetic-induced, rats)
Shortens duration of tetracaine- and
oxybuprocaine-induced corneal anesthesia
[40]
Table 2. Concise summary of the key topical agents trialed to enhance corneal wound healing, along with their mechanisms, representative PubMed citations (PMID/DOI), and evidence levels. Agents trialed for corneal wound healing: mechanisms, evidence, and key references.
Table 2. Concise summary of the key topical agents trialed to enhance corneal wound healing, along with their mechanisms, representative PubMed citations (PMID/DOI), and evidence levels. Agents trialed for corneal wound healing: mechanisms, evidence, and key references.
AgentMechanism/Therapeutic RoleRepresentative ReferenceEvidence Level
Ascorbic acid
(vitamin C)
Antioxidant; promotes collagen synthesis and
stromal repair after chemical injuries
[269]Preclinical
(animal) + small clinical
series
Fibronectin
(autologous)
ECM adhesive protein; supports epithelial
migration and adhesion
[264]Small
uncontrolled clinical series
Hyaluronic acidHydration; enhances epithelial migration and wound closure[133]Controlled
clinical trials
MMP inhibitors (e.g., doxycycline)Inhibit stromal matrix degradation; reduce
corneal melting and perforation risk
[265]Mechanistic +
retrospective
clinical use
EGFStimulates epithelial proliferation and migration[268]Randomized
clinical trial
(historical)
FGF/bFGFPromotes epithelial and stromal repair; used in post-refractive surgery[270]Small clinical studies
NGF
(cenegermin)
Promotes nerve regeneration and corneal healing; approved for neurotrophic ulcers[267]Pivotal randomized controlled trial (RCT); clinical approval
Insulin/IGF-1
(topical)
Stimulates epithelial proliferation;
potential in diabetic/persistent epithelial defects
[242]Emerging RCTs and pilot clinical data
Table 3. Effects of healing agents on corneal ulcer healing, corneal neovascularization, intraocular pressure, and tendon healing (as representative avascular tissue).
Table 3. Effects of healing agents on corneal ulcer healing, corneal neovascularization, intraocular pressure, and tendon healing (as representative avascular tissue).
AgentCorneal Ulcer HealingCorneal
Neovascularization
Intraocular PressureTendon Healing (or Other Avascular Tissue)Notes
BPC 157Most favorable profile, cytoprotection-consistent
AscorbateSimilar to BPC 157
Hyaluronic acidFavorable, cytoprotection-consistent
Insulin0/↓0Close to BPC 157
pattern
Fibronectin? (no direct evidence)Limited data on tendon healing
MMP
inhibitors
Mixed, less favorable
EGF0Divergent ocular vs. tendon effects
FGFIncreased
neovascularization/IOP
NGF00↑ (ligament evidence)Partial, indirect tendon support
IGF-1(Unclear/weak ↑)Problematic, lacks clear corneal evidence
Table 4. Antiangiogenic agents in the context of the cytoprotection “triad”.
Table 4. Antiangiogenic agents in the context of the cytoprotection “triad”.
AgentCorneal Ulcer HealingCorneal
Neovascularization
Intraocular
Pressure (IOP)
Other Avascular Tissues (e.g., Tendon, Skin)
EndostatinEffect on ulcer healing not
determined
↓ (anti-angiogenic)0 (no clear
effect)
↓ (delays in skin/tendon
repair reported)
PAI-1↑ ↓ (pro-angiogenic at physiological levels; anti-angiogenic at higher levels)0 or ↑ (implicated in glaucoma pathogenesis)↓ (impairs
tendon/skin healing)
PEDF↓ (anti-angiogenic)↑ (raises IOP in some models)↑ (supports
tendon/skin healing)
Angiostatin↓ (anti-angiogenic)0 (no clear
effect)
0/↑ (angiostatin-functionalized scaffolds may enhance tendon repair)
TSP-1/2↓ (anti-angiogenic)0 or ↑ (genetic studies suggest possible IOP
elevation)
0/↓ (profibrotic, ECM remodeling, anti-angiogenic; may impair tendon healing)
IFN-α↓ (anti-angiogenic)0 (no clear
effect)
0 (not
determined)
Table 5. Effects of standard anti-neovascularization therapies mapped to the cytoprotection triad.
Table 5. Effects of standard anti-neovascularization therapies mapped to the cytoprotection triad.
AgentCorneal Ulcer HealingCorneal
Neovascularization (CNV)
Intraocular
Pressure (IOP)
Other Avascular Tissues (e.g., Tendon
Healing)
Notes
Corticosteroids↓ (impairs healing)↓ (suppresses CNV)↑ (raises IOP)↓ (delays tendon healing)Effective for CNV but adverse for ulcer healing, IOP, tendon
NSAIDs↓ (impairs healing)↓ (suppresses CNV)↑ (raises IOP)↓ (delays tendon healing)Similar detrimental chain to
corticosteroids
Cyclosporine A↑ (promotes
healing)
↓ (suppresses CNV)↓ (reduces IOP)↓ (impairs tendon healing)More favorable for eye, but tendon effects remain negative
Anti-VEGF drops↑ (via reduced CNV/
inflammation)
↓ (via impaired VEGF-dependent repair)
↓ (suppresses CNV)↑ (raises IOP)↓ (early
inhibition)
↑ (later
recovery)
Dual and context-dependent: beneficial for CNV control but may compromise
repair
Table 6. Effects of standard glaucoma therapies mapped to the cytoprotection triad.
Table 6. Effects of standard glaucoma therapies mapped to the cytoprotection triad.
Drug Class/AgentCorneal
Ulcer
Healing
Corneal
Neovascularization
Intraocular Pressure (IOP) EffectLinked Avascular Tissue Healing (e.g.,
Tendon)
Notes/
Mechanistic
Insight
Alpha 2-agonistsCounteracts IOP but may impair corneal and tendon healing; suggests detrimental cytoprotective profile.
Beta-blockersReduces IOP and supports healing; aligns well with cytoprotection
concept.
Carbonic
Anhydrase (CA) Inhibitors
Lowers IOP; corneal healing impaired but tendon healing supported, possibly via broader
CA-related pathways.
Muscarinic
Agonists (e.g.,
Pilocarpine)
↑/↓Dual effect on corneal healing; supports tendon proliferation; complex
cytoprotective profile.
Rho-kinase
Inhibitors (e.g.,
Ripasudil, Fasudil,
Netarsudil)
↓/↑ (agent-dependent)Reduces IOP, improves corneal healing, supports tendon stem/progenitor mechanics; generally
cytoprotective.
Prostaglandin
Analogues
(Latanoprost)
↑/↓ (preservative-dependent)↓/↑ (preservative-dependent)↑ (low PGE2),
↓ (high PGE2)
Effects variable depending on preservative; dual effects make cytoprotection assessment complex.
Table 7. Grouped agents according to their shared profile within the cytoprotection “triad.” Abbreviations: CNV, corneal neovascularization; IOP, intraocular pressure.
Table 7. Grouped agents according to their shared profile within the cytoprotection “triad.” Abbreviations: CNV, corneal neovascularization; IOP, intraocular pressure.
Shared ProfileAgentsCorneal Ulcer HealingCNVIOPTendon/Skin HealingCytoprotection Alignment
Optimal profile (full triad
alignment)
BPC 157,
Ascorbate, Insulin
Strong; consistent with cytoprotection
Healing +,
CNV –, IOP –,
Tendon +
β-blockers, Rho-kinase inhibitorsFavorable; close to optimal
Healing +,
CNV 0, IOP 0,
Tendon +
NGF00Partial; focused on repair side
Healing +,
CNV –, IOP ↑,
Tendon +
Hyaluronic acid, PEDF Mixed; tendon support but IOP concern (PEDF IOP elevation model-dependent)
Healing +,
CNV –, IOP 0/↑, Tendon –
Cyclosporine A, MMP inhibitors0/↑Partial; corneal support but tendon adverse
Healing +,
CNV +, IOP ↑,
Tendon +/?
EGF, FGF, IGF-1Pro-angiogenic bias; not cytoprotection-consistent
Healing +,
CNV +, IOP ↑,
Tendon ↑
Fibronectin↑ (no direct
evidence)
Pro-angiogenic; tendon repair unclear
Healing +,
CNV –, IOP 0,
Tendon 0/–
Angiostatin, Thrombospondin-1/2, IFN-α00/↓Anti-angiogenic but repair-impaired
Healing –,
CNV –, IOP ↑,
Tendon –
Corticosteroids, NSAIDs,
Anti-VEGF
Anti-CNV but globally
adverse
Healing 0,
CNV –, IOP 0,
Tendon–
Endostatin, PAI-1 (high levels)00Anti-CNV, impairs repair
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

Masnec, S.; Kokot, A.; Kralj, T.; Zlatar, M.; Loncaric, K.; Sablic, M.; Kalauz, M.; Beslic, I.; Oroz, K.; Mrvelj, B.; et al. Challenge of Corneal Ulcer Healing: A Novel Conceptual Framework, the “Triad” of Corneal Ulcer Healing/Corneal Neovascularization/Intraocular Pressure, and Avascular Tendon Healing, for Evaluation of Corneal Ulcer Therapy, Therapy of Neovascularization, Glaucoma Therapy, and Pentadecapeptide BPC 157 Efficacy. Pharmaceuticals 2025, 18, 1822. https://doi.org/10.3390/ph18121822

AMA Style

Masnec S, Kokot A, Kralj T, Zlatar M, Loncaric K, Sablic M, Kalauz M, Beslic I, Oroz K, Mrvelj B, et al. Challenge of Corneal Ulcer Healing: A Novel Conceptual Framework, the “Triad” of Corneal Ulcer Healing/Corneal Neovascularization/Intraocular Pressure, and Avascular Tendon Healing, for Evaluation of Corneal Ulcer Therapy, Therapy of Neovascularization, Glaucoma Therapy, and Pentadecapeptide BPC 157 Efficacy. Pharmaceuticals. 2025; 18(12):1822. https://doi.org/10.3390/ph18121822

Chicago/Turabian Style

Masnec, Sanja, Antonio Kokot, Tamara Kralj, Mirna Zlatar, Kristina Loncaric, Marko Sablic, Miro Kalauz, Iva Beslic, Katarina Oroz, Bozana Mrvelj, and et al. 2025. "Challenge of Corneal Ulcer Healing: A Novel Conceptual Framework, the “Triad” of Corneal Ulcer Healing/Corneal Neovascularization/Intraocular Pressure, and Avascular Tendon Healing, for Evaluation of Corneal Ulcer Therapy, Therapy of Neovascularization, Glaucoma Therapy, and Pentadecapeptide BPC 157 Efficacy" Pharmaceuticals 18, no. 12: 1822. https://doi.org/10.3390/ph18121822

APA Style

Masnec, S., Kokot, A., Kralj, T., Zlatar, M., Loncaric, K., Sablic, M., Kalauz, M., Beslic, I., Oroz, K., Mrvelj, B., Beketic Oreskovic, L., Oreskovic, I., Strbe, S., Staresinic, B., Slivsek, G., Boban Blagaic, A., Seiwerth, S., Skrtic, A., & Sikiric, P. (2025). Challenge of Corneal Ulcer Healing: A Novel Conceptual Framework, the “Triad” of Corneal Ulcer Healing/Corneal Neovascularization/Intraocular Pressure, and Avascular Tendon Healing, for Evaluation of Corneal Ulcer Therapy, Therapy of Neovascularization, Glaucoma Therapy, and Pentadecapeptide BPC 157 Efficacy. Pharmaceuticals, 18(12), 1822. https://doi.org/10.3390/ph18121822

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