Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. May 15, 2025; 16(5): 101509
Published online May 15, 2025. doi: 10.4239/wjd.v16.i5.101509
Therapeutic potential of integrins in diabetic retinopathy
Li-Mei He, Xin-Qi Chen, Department of Internal Medicine, Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
Sarul Borjigin, Zhao-Li Yan, Ming-Jie Wang, Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
ORCID number: Li-Mei He (0009-0004-4894-8877); Sarul Borjigin (0000-0001-6547-5865); Xin-Qi Chen (0009-0007-9145-7442); Zhao-Li Yan (0000-0003-2864-103X); Ming-Jie Wang (0009-0003-9082-9348).
Co-corresponding authors: Zhao-Li Yan and Ming-Jie Wang.
Author contributions: He LM substantially contributed to literature search, interpretation of relevant literature, article drafting, and revision; Borjigin S and Chen XQ contributed to the interpretation of relevant literature, prepared the table and figure, and revised the paper; All authors have read and approved the final manuscript. Yan ZL and Wang MJ contributed to the conception and design of the article, supervised the entire project process, and revised the article. Yan ZL and Wang MJ contributed equally to this study as co-corresponding authors.
Supported by the Natural Science Foundation of Inner Mongolia, No. 2022MS08057; and 2022 Autonomous Region Medical and Health Science and Technology Plan Projects, No. 202202190.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Zhao-Li Yan, MD, Chief Physician, Department of Endocrinology, The Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Road, Hohhot 010000, Inner Mongolia Autonomous Region, China. aliceyzl@126.com
Received: September 18, 2024
Revised: January 17, 2025
Accepted: February 20, 2025
Published online: May 15, 2025
Processing time: 219 Days and 22.4 Hours

Abstract

Diabetic retinopathy (DR), a leading cause of visual loss, is the result of microvascular damage induced by prolonged hyperglycemia. Numerous studies have revealed the pivotal role of integrins in the pathogenesis of DR, particularly in key processes such as inflammation, vascular leakage, microthrombus formation, and angiogenesis. Consequently, targeting integrins is considered a promising strategy for the treatment of DR. This review focuses on the function of integrins in DR and their potential as therapeutic targets. It describes the molecular mechanisms through which integrins influence DR progression and summarizes the latest outcomes of integrin antagonist-based therapeutic strategies in clinical studies, evaluating their efficacy and potential challenges, which offer promise as novel treatment options for DR.

Key Words: Diabetic retinopathy; Integrin; Inflammation; Microvascular permeability; Microthrombus formation; Angiogenesis; Integrin inhibitor

Core Tip: Diabetic retinopathy (DR) is a serious and common complication of diabetes mellitus. Prolonged high glucose levels damage microvasculature in the retina and can lead to blindness. Current treatment options are limited, warranting novel therapeutic strategies. Therapies targeting integrins have shown promise. This review outlines the current understanding of the role of integrins in DR and the findings of preclinical studies and clinical trials investigating the potential of various integrin-targeted therapies. Thus, this review paper captures the current state of the field and identifies possible avenues for further study and treatment approaches.



INTRODUCTION

Diabetes mellitus (DM) is a chronic disease that leads to multi-organ damage through a cascade of homeostatic dysfunctions, resulting in macrovascular and microvascular complications. In 2022, approximately 828 million adults worldwide were estimated to have DM, representing a stark increase of 630 million compared to that in 1990[1]. Among this population, an estimated 35% have coexisting diabetic retinopathy (DR)[2]. From 1990 to 2020, the number of patients with vision-threatening DR has soared from 1.4 million to approximately 3.2 million[3]. Projections indicate that the global prevalence and disease burden of DR will continue to increase in the coming decades, with the patient population anticipated to rise from approximately 103 million in 2020 to 130 million in 2030 and potentially 161 million by 2045[4].

In its early stages, DR is typically asymptomatic, causing most patients to present with advanced disease and severe retinal damage, complicating treatment and worsening prognosis. Current DR therapies, including laser photocoagulation, intravitreal corticosteroid injection, and anti-vascular endothelial growth factor (VEGF) therapy, are effective but have notable limitations. Anti-VEGF therapy demonstrates therapeutic effects predominantly in the advanced stages of DR and necessitates frequent intraocular injections, posing infection risks and other potential side effects. Moreover, up to 50% of patients exhibit limited response to anti-VEGF monotherapy[5,6]. Prolonged corticosteroid use can significantly impact the eye, inducing complications such as elevated intraocular pressure and glaucoma[5]. Laser treatment can damage healthy retinal tissue, restrict visual fields, and impair night vision[7].

Elucidating the pathological mechanisms underlying DR has facilitated the development of innovative therapeutic strategies. Among the diverse biomolecules and cellular pathways involved in DR pathogenesis, therapies targeting integrins have shown promise. This class of transmembrane glycoprotein receptors, mediate interactions between cells and the extracellular matrix (ECM) and regulate cell proliferation, differentiation, and migration[8]. DR pathological processes are intricately linked to the aberrant proliferation and migration of vascular endothelial cells, as well as dysregulated neovascularization[9]. Inhibiting integrin activity may slow DR progression and address certain limitations of current treatments, such as altering the route of administration and reducing the need for frequent intraocular injections. This will alleviate patients pain while improving treatment convenience and compliance. Nonetheless, research on this strategy remains in its early stages, and further clinical studies are required to confirm its safety and efficacy. Elucidating the relationship between retinal integrin expression and the pathological alterations that occur in DR, including inflammatory responses, vascular permeability, microthrombus formation, and neovascularization, as well as potential interventional strategies, could provide novel insights for DR treatment.

OVERVIEW OF INTEGRINS
Composition and classification of integrins

Integrins are composed of two distinct subunits, α and β, which form heterodimers through non-covalent interactions. Each subunit consists of extracellular, transmembrane, and intracellular domains. The extracellular region of the α subunit comprises β-propeller, Thigh, Calf-1, and Calf-2 domains. Notably, the α1, α2, α10, α11, αD, αX, αM, and αL subunits contain a I domain inserted between blades two and three of the seven-bladed β-propeller. The extracellular domain of the β subunit is more complex, comprising βI, Hybrid, plexin-semaphorin-integrin, four epidermal growth factor, and β-tail domains (Figure 1). In vertebrates, 18 α subunits and 8 β subunits have been identified, which combine to form 24 different integrins[8,10]. Based on structural similarities and ligand specificity, the integrin receptor family is categorized into the following four classes: (1) Receptors recognizing the Arg-Gly-Asp (RGD) sequence; (2) Laminin receptors; (3) Collagen receptors; and (4) Leukocyte-specific receptors[8].

Figure 1
Figure 1 Schematic illustration of integrin conformations. A: Bent-closed, inactive; B: Extended-closed, transition between inactive and active; integrins are extended but the ligand-binding site is not exposed; C: Extended-open, active; integrins are fully extended and the ligand-binding site is exposed. EGF: Epidermal growth factor; PSI: Plexin-semaphorin-integrin. Created by figdraw.com (Supplementary material).
Integrin signaling

Integrins exist in three conformational states: Bent-closed, where both subunits are folded and maintained in an inactive state by endogenous inhibitory proteins; extended-closed, following inside-out (by the binding of activators to the intracellular domain) or outside-in (by the binding of extracellular ligands) activation to an intermediate state, with extension of the integrin but no exposure of the ligand-binding site; and extended-open, the active state achieved following simultaneous binding of the actin cytoskeleton and extracellular ligands, with full extension and exposure of the ligand-binding site[11,12] (Figure 1). Integrins are crucial for stable cell–ECM adhesion and signal transduction, binding specific ECM ligands and transmitting signals bidirectionally across the membrane[12]. Integrins’ intracellular domains interact with cytoskeletal proteins such as talin, facilitating inside-out signaling and conformational changes. Although integrins lack intrinsic enzymatic activity, they mediate outside-in signaling by activating downstream pathways, including phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB/AKT), extracellular signal-regulated kinase (ERK), and mitogen-activated protein kinase. Integrins can therefore function as adhesion molecules and signaling receptors, recruiting intracellular adaptors, cytoskeletal proteins, and signaling molecules to their cytoplasmic domains to achieve signal transduction[12,13].

COMPOSITION AND FUNCTION OF INTEGRINS EXPRESSED IN THE RETINA

The retina is transparent, thin, and composed of ten distinct layers. The integrin subunits α2, α3, α4, α5, and β2 are expressed in most regions of the human retina[8]. The retinal pigment epithelium, which is crucial for maintaining retinal health and visual function, ensures precise alignment of photoreceptor outer segments and the efficient phagocytosis of shed outer segment fragments, processes that are dependent on the specific distribution of integrin αVβ5 on its apical surface[14]. Additionally, integrin β8 is highly expressed in mouse Müller glial cells and retinal ganglion cells; loss of this integrin significantly impairs the normal development of the mouse retinal system[15].

ROLE OF INTEGRINS IN DR
Inflammation

Integrin involvement in inflammatory cell activation: DR is a chronic inflammatory condition, in which integrins are involved in activating inflammatory cells, inducing leukocyte adhesion and aggregation within the bloodstream and monocyte-macrophage infiltration. Leukocytes are rapidly recruited to the inflammation site through interactions with vascular endothelial cells. Integrin antagonists reduce leukocyte recruitment by decreasing the expression of integrins in their active form, disrupting leukocyte–endothelial cell adhesion, and transendothelial migration[16]. In DR, chronic inflammatory responses exacerbate retinal microvascular damage. Damaged tissues release signaling molecules, such as cytokines and chemokines, prompting endothelial cell activation. Upon receiving chemotactic signals, leukocytes adhere loosely and roll along the vessel wall via selectins. Chemokines on the endothelial surface activate leukocytes, leading to integrin activation and firm adhesion between leukocytes and endothelial cells. Subsequently, leukocytes change shape and migrate through the endothelial layer into the damaged tissue, a process mediated by complex interactions between leukocytes and endothelial cells[17]. Endothelial cells express various adhesion molecules, such as vascular cell adhesion molecule-1 (VCAM-1 or CD106) and intercellular adhesion molecule-1 (ICAM-1 or CD54), to facilitate interactions with leukocytes. Activated leukocyte surface integrins strongly adhere to these ligands, ultimately allowing transendothelial migration into inflammatory tissues. For example, the binding of integrin αLβ2 to ICAM-1 enhances the adhesion of neutrophils to the vascular wall, whereas the interactions between α4β1 and the RGD-binding integrin αVβ3 with VCAM-1 enable lymphocytes to rapidly migrate across the endothelium into sites of inflammation[17,18]. Cysteine-rich 61, a pro-angiogenic factor elevated in DR[19], induces monocyte chemoattractant protein-1 (MCP-1) expression through the combined action of integrin αVβ3, focal adhesion kinase (FAK), and the PI3K/AKT and IκB kinase/nuclear factor (NF)-κB signaling pathways, promoting macrophage infiltration[20].

Interactions between integrins and inflammatory factors: The levels of various inflammatory factors, including but not limited to IL-1β, tumor necrosis factor (TNF)-α, MCP-1, and transforming growth factor (TGF)-β1, are elevated in the ocular fluid of patients with DR. These factors interact with integrins to modulate a range of inflammatory processes[21].

Integrin αVβ3 influences macrophage functions by activating signaling pathways such as PI3K/AKT, protein kinase C, and ERK. Activation of integrin αVβ3 enhances NF-κB activity, promoting the expression of pro-inflammatory genes and the production of inflammatory mediators like TNF-α. Signaling pathway regulation can therefore alter macrophage responses, providing potential strategies for the treatment of inflammatory diseases[22]. MCP-1, also known as CCL2, is found at higher levels in the vitreous humor than in the serum of patients with DR, indicating local ocular expression[23]. MCP-1 activates β1 integrin, thereby promoting the adhesion of THP-1 monocyte-like cells, primarily through the ERK signaling pathway. Inhibition of the upstream MEK pathway can suppress this effect[24].

TGF-β is a multifaceted growth factor that is pivotal in development and immunity under physiological conditions. The gene products of TGF-β encompass TGF-β1, -β2, and -β3 and exist in a latent form known as latency-associated peptide (LAP)-TGF-β. TGF-β is expressed in various cells, including macrophages, dendritic cells, and lymphocytes. Notably, the LAP of TGF-β1 and TGF-β3 contains integrin-binding motifs RGDLXXL/I, enabling them to bind to integrins αVβ6 and αVβ8. Through autocrine or paracrine signaling, these bound TGF-β isoforms regulate cell proliferation, differentiation, and activation. They also modulate the expression of adhesion molecules, facilitating the chemotaxis of associated inflammatory cells[25,26]. Gerhardinger et al[27] demonstrated the significance of the TGF-β signaling pathway in a rat model of DR, revealing significant activation of the TGF-β pathway and the differential expression of 20 TGF-β-related genes. This was associated with oxidative stress, inflammation, vascular remodeling, and apoptosis in retinal vessels. Moreover, aldose reductase inhibitors such as sorbitol and aspirin effectively inhibit TGF-β pathway upregulation, indicating their potential value in DR prevention and treatment.

Microvascular permeability

VEGF plays a key role in regulating angiogenesis, exhibiting potent vasodilatory effects, and enhancing vascular permeability after binding to two tyrosine kinase receptors: VEGF receptor (VEGFR)1 and VEGFR2[28]. AXT-107 is a collagen IV-derived peptide capable of binding αVβ3 and α5β1 integrins. In human retinal endothelial cells, AXT-107 inhibits integrin β3 by interfering with the co-clustering of VEGFR-2 and β3 integrin while simultaneously reducing the overall level of VEGFR2 through increased internalization, ubiquitination, and degradation. VEGF-induced vascular leakage was reduced by 86%, one month after injection of AXT-107 into rabbit eyes[29] (Table 1). Consequently, decreasing the activity of integrin β3 may reduce pathological vascular leakage.

Table 1 Summary of integrin antagonists presented in the text.
Drug name
Target integrin receptor(s)
Stage of research
Route of administration
Effect
Ref.
AXT-107αVβ3, α5β1Clinical researchIntravitreal injectionReduces leakage, neovascularization and inflammation[29,73]
JNJ-26076713αVβ3, αVβ5Preclinical research/Reduces leakage, neovascularization and inflammation[32]
ATN-161α5β1Preclinical research/Reduces leakage, neovascularization and inflammation[41]
RisuteganibαVβ3, αVβ5, α5β1, αMβ2Clinical researchIntravitreal injectionReduces leakage, inflammation and angiogenesis, improves mitochondrial function [5,56,57]
THR-687αVβ3, αVβ5, α5β1Clinical researchIntravitreal injectionReduces leakage and fibrosis[58-60]
SF-0166αVβ3, αVβ6, αVβ8Clinical researchTopical dropInhibits neovascularization, decreases lesion area and reduces leakage[61,62,74]
OTT166αVβ3, αVβ6, αVβ8Clinical researchTopical dropReduces inflammation, fibrosis, leakage, and neovascularization[63,75]
RUC-4αIIbβ3Clinical researchSubcutaneous injectionInhibits thrombus formation[71,72,76]

Angiopoietin-like proteins (ANGPTLs) regulate angiogenesis and share similarities with angiopoietins. Among them, angiopoietin-like protein 4 (ANGPTL4) reduces hypoxia-induced vascular permeability, suggesting its potential therapeutic value in preventing or treating hypoxia-related diseases such as DR. Co-administration of ANGPTL4 with a blocking antibody against αVβ3 integrin abolishes this effect, indicating that ANGPTL4 exerts its protective actions by interacting with αVβ3 integrin. Mechanistically, its binding to integrin αVβ3 enhances Src kinase recruitment, inhibiting downstream VEGFR2 signaling; this stabilizes blood vessels, increases tight junction integrity, and resists hypoxia-induced increases in vascular permeability[30]. Therefore, novel drugs targeting VEGFR2 signaling can reduce vascular permeability by increasing ANGPTL4 Levels or modulating its interaction with integrin αVβ3, improving therapeutic outcomes.

In studies on sepsis and acute lung injury, integrin αVβ3 inhibition or knockout increases vascular leakage[31]. However, administering the integrin αV antagonist JNJ-26076713 to rats with DM significantly inhibits retinal vascular permeability[32] (Table 1). These contradictory findings may be due to differences in models or mechanisms, requiring further research on integrin targeting therapies.

Pericyte loss is a hallmark of early-stage DR[33]. Pericytes provide structural support to capillaries, and their loss leads to localized capillary wall leakage. Both in vitro and in vivo studies have shown that high glucose levels induce pericyte apoptosis[34,35]. ANGPT2 expression level is upregulated in patients with DR[36]. Park et al[37] confirmed that, in mice with STZ-induced DM, ANGPT2 induces pericyte apoptosis via the p53 pathway under high-glucose conditions, mediated by binding to integrin α3β1. This involves phosphorylation of p53 at Ser-15 through the ERK1/2 pathway, which does not induce apoptosis under normoglycemic conditions. Therefore, controlling blood glucose levels or blocking ANGPT2/integrin signaling may prevent early DR pericyte loss. Elevated ANGPT levels can also induce retinal leakage and astrocyte loss in diabetic mice. In a hyperglycemic environment, ANGPT2 triggers astrocyte apoptosis via the integrin αVβ5/GSK-3β/β-catenin signaling cascade. Notably, inhibition of integrin αVβ5 substantially mitigates apoptosis and vascular leakage[38]. Furthermore, ANGPT2 mediates endothelial cell layer destabilization by activating integrin α5β1, leading to increased vascular leakage[39]. Administering an integrin β1 antibody under inflammatory conditions can improve endothelial cell junction integrity, thereby reducing vascular permeability[40]. Additionally, the integrin α5β1 inhibitor ATN-161 can inhibit NLRP3 inflammasome signaling in the mouse retina, reducing VEGF-induced vascular leakage[41] (Table 1). These findings suggest potential therapeutic strategies and targets for DR.

Prothrombotic state and microthrombosis

In patients with DM, metabolic disorders disrupt their physiological balance, resulting in a prothrombotic state characterized by platelet hypersensitivity[42]. Integrin αIIbβ3, the principal surface receptor for platelet adhesion to the ECM, mediates platelet aggregation by binding plasma fibrinogen. Under normal conditions, a complex positive feedback loop is maintained, with nitric oxide and prostaglandin I2 counteracting platelet activation and aggregation mediated by plasma thrombin, platelet-derived growth factor, and adenosine diphosphate. However, type 2 DM disrupts this balance; in vivo, platelet aggregation increases under insulin resistance, suggesting that platelets are less responsive to nitric oxide and prostaglandin I2[43]. Consequently, patients with type 2 DM may be at increased risk of thrombosis[42,44].

Platelets from patients with DR exhibit heightened sensitivity to low concentrations of adenosine diphosphate and an increased propensity to aggregate in vitro[45]. If this is reflected in vivo, microthrombosis and vascular obstruction may occur. DR pathological changes include retinal microvascular occlusion, which may result from luminal narrowing caused by endothelial cell proliferation or basement membrane thickening, or from emboli formed by aggregates of platelets, fibrin, and cholesterol[46]. Ju et al[47] established a mouse model of thrombi formation and showed that, compared to the non-diabetic control group mice, diabetic mice exhibited a significant increase in the platelet aggregation rate, aggregation extent, and thrombus area. Pretreatment with an integrin αIIbβ3 antagonist or a PI3K inhibitor suppressed platelet aggregation, whereas pretreatment with the conventional antiplatelet drugs aspirin and clopidogrel did not. This demonstrates the complexity of managing the clinical risk of thrombus formation in patients with DM.

The activation of integrins αVβ3 and αIIbβ3 can be mediated through the allosteric binding of inflammatory chemokines such as CCL5, CX3CL1, and CXCL12, through a mechanism independent of the classical inside-out signaling pathway. The binding of these chemokines enhances the affinity of integrins for the ECM, thereby triggering platelet aggregation and potentially thrombosis[48]. Niu et al[49] demonstrated that the Src family kinase inhibitor PP2 and the PI3K inhibitor wortmannin inhibit integrin αIIbβ3 outside-in signaling-induced shape changes and platelet aggregation. A study by Qin et al[50] using a mouse model of DM emphasized the importance of integrin αIIbβ3 in the early stages of thrombosis. Notably, under diabetic conditions, injured endothelial cells release endothelial microparticles containing protein disulfide isomerase, which accelerates platelet activation by binding to integrin αIIbβ3. Inhibiting protein disulfide isomerase mitigates this process, suggesting a potential therapeutic strategy for DM-related thrombotic diseases. Therefore, a deeper understanding of platelet function and the role of integrin αIIbβ3 may improve management of DM.

Angiogenesis

The high metabolic demands of the retina make it particularly susceptible to oxidative stress and metabolic dysregulation, which can initiate pathological neovascularization and tissue damage.

The complexity of angiogenesis is demonstrated by the regulatory role of integrins in the Ras-ERK pathway, with αVβ3 antibody inhibiting basic fibroblast growth factor-mediated Ras activity and c-Raf activation, whereas αVβ5 antibody affected VEGF-mediated Ras activity[51]. Upon VEGF activation, integrin αVβ5 on the cell surface is mobilized, promoting cell proliferation, migration, and invasion of tissues. FAK serves as the first signaling molecule in the integrin-mediated signal transduction system, forming a FAK/Src/αVβ5 complex, which subsequently activates Ras–Raf–ERK step-wise, ultimately promoting cell proliferation through the ERK1/2 pathway[51,52]. In contrast, bFGF activates c-Raf through αVβ3, FAK, and PAK downstream of Ras[51]. Alternatively, research indicates that VEGF activates VEGFR-2, which in turn induces the activation of c-Src and results in the phosphorylation of β3 integrin. Concurrently, the tyrosine phosphorylation of β3 integrin provides feedback to further enhance VEGFR-2 activation. This cross-activation mechanism has opened up novel avenues for anti-angiogenic strategies[53]. The distinct modes of integrin action result in varied cellular responses and angiogenic pathways. In early quail embryo vascular development, treatment with anti-αVβ3 antibodies can block vascular formation[54], and in mice, pharmacological or genetic disruption of β3 integrin inhibits the coverage of retinal vascular smooth muscle cells and arterial maturation[55]. These results underscore the critical role of β3 integrin in vascular maturation and stability, the complexity of the cellular mechanisms behind angiogenesis, and the importance of precise interventions. Understanding the specific roles of integrins and their associated signaling cascades provides avenues for the development of targeted therapies that can more effectively address DR-associated vascular abnormalities.

ANTI-INTEGRIN THERAPIES

Risuteganib is an innovative, vitreous-injected synthetic RGD oligopeptide that primarily inhibits four integrins: αVβ3, αVβ5, α5β1, and αMβ2[5]. The DEL MAR 2b trial (NCT02348918) explored the use of risuteganib as a follow-up therapy after a single anti-VEGF injection in patients with diabetic macular edema (DME)[5]. Compared to anti-VEGF monotherapy, sequential therapy demonstrated comparable efficacy to bevacizumab monotherapy in terms of the primary endpoint and required fewer total injections, with sustained efficacy observed for 12 weeks after the last risuteganib dose. Furthermore, risuteganib exhibited promising efficacy in patients with an inadequate response to anti-VEGF therapy[5,56,57] (Table 1).

THR-687, an innovative integrin receptor antagonist, has the ability to tightly bind to integrins αVβ3, αVβ5, and α5β1 at extremely low nanomolar concentrations, thereby exerting a significant impact on crucial pathological processes such as cell migration and proliferation[58]. In a Phase I clinical trial (NCT03666923) conducted on patients with DME, THR-687 exhibited favorable safety and tolerability profiles across all tested dose levels[58,59]. Following administration, the mean best-corrected visual acuity (BCVA) increased by 7.2 Letters on day 7 after injection, peaking at a mean improvement of 9.2 Letters one month later and sustaining an overall mean BCVA improvement of 8.3 Letters up to three months[58]. Furthermore, data from Part A of a Phase II trial (NCT05063734) for THR-687 in DME underscored the drug’s safety and tolerability. However, despite the encouraging safety profile, evidence of efficacy at key endpoints, namely BCVA and central subfield thickness, remains insufficient[60]. Hence, THR-687, as a novel integrin receptor antagonist, has demonstrated promising safety and potential therapeutic efficacy in treating DME (Table 1).

SF-0166 is a small molecule inhibitor of integrin αVβ3 that also inhibits αVβ6 and αVβ8 in mice. Topical application of SF-0166 can suppress neovascularization, with a reduction in lesion area comparable to that achieved by intravitreal bevacizumab injection; the drug concentration can be maintained for over 12 hours[61]. Given its ability to inhibit neovascularization induced by VEGF and other growth factors, SF-0166 offers advantages over VEGF-only antagonists. A Phase I/II, randomized, double-blind, multicenter clinical trial (NCT02914613) enrolled 40 patients with DME and randomly assigned them to two dose groups (2.5% or 5%). Patients self-administered SF-0166 ophthalmic solution topically twice daily for twenty-eight days. SF-0166 exhibited biological activity in both dose groups, with 53% of patients experiencing reduced retinal thickness and improved visual acuity. In terms of safety, the trial achieved its primary endpoint, with mild ocular adverse events occurring in six patients, one of which may have been related to the drug[62] (Table 1). In summary, SF-0166 has shown promising efficacy in inhibiting neovascularization and reducing retinal lesions in both preclinical and clinical studies. Its safety in patients with DME has also been verified, demonstrating significant biological activity and sustained therapeutic effects, which support further clinical research.

OTT166 is a highly potent and selective small molecule RGD-binding integrin inhibitor targeting integrins αVβ3, αVβ6, and αVβ8[8]. In a multicenter, randomized, double-blind Phase II study of DR: Early Active Management (NCT05409235), enrolled 225 adult patients with moderate to severe non-proliferative DR or mild proliferative DR and mild visual impairment. Patients were randomly assigned to four groups: 5% OTT166 topical eyedrops twice or four times daily via self-administration, placebo eyedrops twice or four times daily via self-administration for 24 weeks. Although OTT166 (5%) demonstrated good safety performance, it failed to show statistically significant improvement in the primary or key secondary efficacy endpoints. Nevertheless, safety remains a notable advantage, providing a foundation for further clinical trials[63] (Table 1).

Platelet integrin αIIbβ3 inhibitors block the final pathway of platelet activation, preventing platelet aggregation[64]. Intracoronary injection of the integrin αIIbβ3 antagonist abciximab, approved for clinical use in 1994[65,66], can significantly reduce the incidence of major cardiovascular events within one year in patients with DM and ST-segment elevation myocardial infarction[67,68]. However, oral integrin αIIbβ3 inhibitors have not been approved due to the increased risk of bleeding and mortality[69,70]. The new integrin αIIbβ3 antagonist, RUC-4, is administered subcutaneously and binds to the metal ion-binding site of integrin β3, blocking its interaction with fibrinogen and preventing conformational changes. RUC-4 has advanced to clinical trials, demonstrating its safety and tolerability in the studied populations. However, the efficacy and safety of RUC-4 in patients with DR remain unexplored, necessitating further clinical studies to ascertain its potential therapeutic benefits within this patient cohort[71,72] (Table 1).

CONCLUSION

DR, a common and complex complication of DM, involves a myriad of interdependent biological processes, exemplified by the multifaceted roles of integrins in DR. This review summarizes the contributions of integrins to the pathogenesis of DR, particularly their roles in inflammation, microvascular permeability, thrombosis, microthrombus formation, and neovascularization. Although we have recognized the role of integrins in promoting DR, their molecular mechanisms require further characterization, especially regarding the crucial role of integrins in diabetic vascular dysfunction. The ability of integrins to act upstream and downstream of the VEGF pathway accounts for why some patients with poor responses to anti-VEGF therapy experience good efficacy with risuteganib[5]. Therefore, risuteganib holds promise as a new option in the treatment of DME, particularly in reducing the number of required injections and improving efficacy in refractory patients. Furthermore, although OTT166 failed to significantly improve the primary endpoint in the overall patient population in a Phase II clinical trial[63], the effect observed in patients with more severe baseline lesions warrants further investigation. Additionally, more precise patient selection must be implemented to assess OTT166 efficacy in these specific patient groups. Optimizing the therapeutic effect of OTT166 can be further achieved by increasing the dosage, extending the treatment duration, or combining it with other treatment methods. Non-invasive treatment options can provide convenient therapy for patients, delaying or reversing DR progression and changing the management approach to DR. Although anti-integrin therapy holds potential for treating DME and DR, its efficacy is variable. For instance, patients who exhibit poor responses to VEGF inhibitors demonstrate better reactions to anti-integrin therapy[5], suggesting that anti-integrin treatment may be more suitable for specific patient populations rather than all patients with DME. Therefore, further research is required to identify which patients are most likely to benefit. Current clinical trials primarily focus on short-term efficacy, whereas the long-term effects and safety of anti-integrin therapy have not been fully evaluated. Considering that integrins participate in various physiological and pathological processes, their inhibitors may affect multiple pathways. Consequently, long-term exposure to integrin inhibitors may produce unknown side effects on the eyes and other organs, necessitating long-term follow-up studies. Moreover, even with fewer injections, intravitreal injection remains a relatively invasive treatment, which may affect patient adherence. Therefore, the development of more convenient and patient-friendly drug administration methods represents an important research direction. Future research should concentrate on elucidating the specific functionalities and underlying mechanisms of integrins at various stages of DR, examining how different integrin subtypes interact and synergize. This will help resolve the associated intricate cellular signaling networks and determine whether improving the hypercoagulable state can be achieved in patients with diabetes when blood glucose levels are adequately managed and controlled. DR treatment remains challenging, yet integrins and their associated signaling pathways offer numerous potential therapeutic targets. Further research on the molecular mechanisms involved should facilitate the development of more precise and effective treatments.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade B, Grade C

Novelty: Grade B, Grade B

Creativity or Innovation: Grade B, Grade D

Scientific Significance: Grade B, Grade B

P-Reviewer: Carbone F; Luo B; Pappachan JM; Sun XD S-Editor: Li L L-Editor: A P-Editor: Xu ZH

References
1.  NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in diabetes prevalence and treatment from 1990 to 2022: a pooled analysis of 1108 population-representative studies with 141 million participants. Lancet. 2024;404:2077-2093.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 73]  [Reference Citation Analysis (0)]
2.  Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, Colagiuri S, Guariguata L, Motala AA, Ogurtsova K, Shaw JE, Bright D, Williams R; IDF Diabetes Atlas Committee. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes Res Clin Pract. 2019;157:107843.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5345]  [Cited by in RCA: 5663]  [Article Influence: 943.8]  [Reference Citation Analysis (8)]
3.  Flaxman SR, Bourne RRA, Resnikoff S, Ackland P, Braithwaite T, Cicinelli MV, Das A, Jonas JB, Keeffe J, Kempen JH, Leasher J, Limburg H, Naidoo K, Pesudovs K, Silvester A, Stevens GA, Tahhan N, Wong TY, Taylor HR; Vision Loss Expert Group of the Global Burden of Disease Study. Global causes of blindness and distance vision impairment 1990-2020: a systematic review and meta-analysis. Lancet Glob Health. 2017;5:e1221-e1234.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1508]  [Cited by in RCA: 2006]  [Article Influence: 250.8]  [Reference Citation Analysis (0)]
4.  Tan TE, Wong TY. Diabetic retinopathy: Looking forward to 2030. Front Endocrinol (Lausanne). 2022;13:1077669.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 112]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
5.  Shaw LT, Mackin A, Shah R, Jain S, Jain P, Nayak R, Hariprasad SM. Risuteganib-a novel integrin inhibitor for the treatment of non-exudative (dry) age-related macular degeneration and diabetic macular edema. Expert Opin Investig Drugs. 2020;29:547-554.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
6.  Dulull N, Kwa F, Osman N, Rai U, Shaikh B, Thrimawithana TR. Recent advances in the management of diabetic retinopathy. Drug Discov Today. 2019;24:1499-1509.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
7.  Stewart MW. Treatment of diabetic retinopathy: Recent advances and unresolved challenges. World J Diabetes. 2016;7:333-341.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 117]  [Cited by in RCA: 104]  [Article Influence: 11.6]  [Reference Citation Analysis (2)]
8.  Van Hove I, Hu TT, Beets K, Van Bergen T, Etienne I, Stitt AW, Vermassen E, Feyen JHM. Targeting RGD-binding integrins as an integrative therapy for diabetic retinopathy and neovascular age-related macular degeneration. Prog Retin Eye Res. 2021;85:100966.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
9.  Wei L, Sun X, Fan C, Li R, Zhou S, Yu H. The pathophysiological mechanisms underlying diabetic retinopathy. Front Cell Dev Biol. 2022;10:963615.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 29]  [Reference Citation Analysis (0)]
10.  Pan L, Zhao Y, Yuan Z, Qin G. Research advances on structure and biological functions of integrins. Springerplus. 2016;5:1094.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 55]  [Cited by in RCA: 62]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
11.  Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10:9-22.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2765]  [Cited by in RCA: 2780]  [Article Influence: 185.3]  [Reference Citation Analysis (0)]
12.  Cabodi S, Di Stefano P, Leal Mdel P, Tinnirello A, Bisaro B, Morello V, Damiano L, Aramu S, Repetto D, Tornillo G, Defilippi P. Integrins and signal transduction. Adv Exp Med Biol. 2010;674:43-54.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 96]  [Cited by in RCA: 97]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
13.  Shattil SJ, Newman PJ. Integrins: dynamic scaffolds for adhesion and signaling in platelets. Blood. 2004;104:1606-1615.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 406]  [Cited by in RCA: 406]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
14.  Mallavarapu M, Finnemann SC. Neural retina and MerTK-independent apical polarity of alphavbeta5 integrin receptors in the retinal pigment epithelium. Adv Exp Med Biol. 2010;664:123-131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
15.  Arnold TD, Ferrero GM, Qiu H, Phan IT, Akhurst RJ, Huang EJ, Reichardt LF. Defective retinal vascular endothelial cell development as a consequence of impaired integrin αVβ8-mediated activation of transforming growth factor-β. J Neurosci. 2012;32:1197-1206.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 56]  [Cited by in RCA: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
16.  Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res. 2021;89:1619-1626.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 85]  [Cited by in RCA: 195]  [Article Influence: 48.8]  [Reference Citation Analysis (0)]
17.  Mrugacz M, Bryl A, Falkowski M, Zorena K. Integrins: An Important Link between Angiogenesis, Inflammation and Eye Diseases. Cells. 2021;10:1703.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
18.  Imhof BA, Weerasinghe D, Brown EJ, Lindberg FP, Hammel P, Piali L, Dessing M, Gisler R. Cross talk between alpha(v)beta3 and alpha4beta1 integrins regulates lymphocyte migration on vascular cell adhesion molecule 1. Eur J Immunol. 1997;27:3242-3252.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 48]  [Cited by in RCA: 49]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
19.  Xiang ZY, Chen SL, Qin XR, Lin SL, Xu Y, Lu LN, Zou HD. Changes and related factors of blood CCN1 levels in diabetic patients. Front Endocrinol (Lausanne). 2023;14:1131993.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
20.  You JJ, Yang CH, Yang CM, Chen MS. Cyr61 induces the expression of monocyte chemoattractant protein-1 via the integrin ανβ3, FAK, PI3K/Akt, and NF-κB pathways in retinal vascular endothelial cells. Cell Signal. 2014;26:133-140.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 33]  [Cited by in RCA: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
21.  Takada YK, Yu J, Fujita M, Saegusa J, Wu CY, Takada Y. Direct binding to integrins and loss of disulfide linkage in interleukin-1β (IL-1β) are involved in the agonistic action of IL-1β. J Biol Chem. 2017;292:20067-20075.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
22.  Antonov AS, Antonova GN, Munn DH, Mivechi N, Lucas R, Catravas JD, Verin AD. αVβ3 integrin regulates macrophage inflammatory responses via PI3 kinase/Akt-dependent NF-κB activation. J Cell Physiol. 2011;226:469-476.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 83]  [Cited by in RCA: 98]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
23.  Mitamura Y, Takeuchi S, Matsuda A, Tagawa Y, Mizue Y, Nishihira J. Monocyte chemotactic protein-1 in the vitreous of patients with proliferative diabetic retinopathy. Ophthalmologica. 2001;215:415-418.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 61]  [Cited by in RCA: 67]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
24.  Ashida N, Arai H, Yamasaki M, Kita T. Differential signaling for MCP-1-dependent integrin activation and chemotaxis. Ann N Y Acad Sci. 2001;947:387-389.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 17]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
25.  Callan A, Jha S, Valdez L, Baldado L, Tsin A. TGF-β Signaling Pathways in the Development of Diabetic Retinopathy. Int J Mol Sci. 2024;25:3052.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
26.  Dong X, Hudson NE, Lu C, Springer TA. Structural determinants of integrin β-subunit specificity for latent TGF-β. Nat Struct Mol Biol. 2014;21:1091-1096.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 93]  [Cited by in RCA: 119]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
27.  Gerhardinger C, Dagher Z, Sebastiani P, Park YS, Lorenzi M. The transforming growth factor-beta pathway is a common target of drugs that prevent experimental diabetic retinopathy. Diabetes. 2009;58:1659-1667.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 64]  [Cited by in RCA: 54]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
28.  Ferrara N. Role of vascular endothelial growth factor in regulation of physiological angiogenesis. Am J Physiol Cell Physiol. 2001;280:C1358-C1366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 723]  [Cited by in RCA: 723]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
29.  Silva RLE, Kanan Y, Mirando AC, Kim J, Shmueli RB, Lorenc VE, Fortmann SD, Sciamanna J, Pandey NB, Green JJ, Popel AS, Campochiaro PA. Tyrosine kinase blocking collagen IV-derived peptide suppresses ocular neovascularization and vascular leakage. Sci Transl Med. 2017;9:eaai8030.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 35]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
30.  Gomez Perdiguero E, Liabotis-Fontugne A, Durand M, Faye C, Ricard-Blum S, Simonutti M, Augustin S, Robb BM, Paques M, Valenzuela DM, Murphy AJ, Yancopoulos GD, Thurston G, Galaup A, Monnot C, Germain S. ANGPTL4-αvβ3 interaction counteracts hypoxia-induced vascular permeability by modulating Src signalling downstream of vascular endothelial growth factor receptor 2. J Pathol. 2016;240:461-471.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 34]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
31.  Su G, Atakilit A, Li JT, Wu N, Bhattacharya M, Zhu J, Shieh JE, Li E, Chen R, Sun S, Su CP, Sheppard D. Absence of integrin αvβ3 enhances vascular leak in mice by inhibiting endothelial cortical actin formation. Am J Respir Crit Care Med. 2012;185:58-66.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 69]  [Cited by in RCA: 71]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
32.  Santulli RJ, Kinney WA, Ghosh S, Decorte BL, Liu L, Tuman RW, Zhou Z, Huebert N, Bursell SE, Clermont AC, Grant MB, Shaw LC, Mousa SA, Galemmo RA Jr, Johnson DL, Maryanoff BE, Damiano BP. Studies with an orally bioavailable alpha V integrin antagonist in animal models of ocular vasculopathy: retinal neovascularization in mice and retinal vascular permeability in diabetic rats. J Pharmacol Exp Ther. 2008;324:894-901.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 45]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
33.  Zhou J, Chen B. Retinal Cell Damage in Diabetic Retinopathy. Cells. 2023;12:1342.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 21]  [Cited by in RCA: 31]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
34.  Naruse K, Nakamura J, Hamada Y, Nakayama M, Chaya S, Komori T, Kato K, Kasuya Y, Miwa K, Hotta N. Aldose reductase inhibition prevents glucose-induced apoptosis in cultured bovine retinal microvascular pericytes. Exp Eye Res. 2000;71:309-315.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 55]  [Cited by in RCA: 63]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
35.  Romeo G, Liu WH, Asnaghi V, Kern TS, Lorenzi M. Activation of nuclear factor-kappaB induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes. Diabetes. 2002;51:2241-2248.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 257]  [Cited by in RCA: 295]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
36.  Cai J, Kehoe O, Smith GM, Hykin P, Boulton ME. The angiopoietin/Tie-2 system regulates pericyte survival and recruitment in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2008;49:2163-2171.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 104]  [Cited by in RCA: 113]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
37.  Park SW, Yun JH, Kim JH, Kim KW, Cho CH, Kim JH. Angiopoietin 2 induces pericyte apoptosis via α3β1 integrin signaling in diabetic retinopathy. Diabetes. 2014;63:3057-3068.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 104]  [Cited by in RCA: 106]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
38.  Yun JH, Park SW, Kim JH, Park YJ, Cho CH, Kim JH. Angiopoietin 2 induces astrocyte apoptosis via αvβ5-integrin signaling in diabetic retinopathy. Cell Death Dis. 2016;7:e2101.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 41]  [Cited by in RCA: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
39.  Hakanpaa L, Sipila T, Leppanen VM, Gautam P, Nurmi H, Jacquemet G, Eklund L, Ivaska J, Alitalo K, Saharinen P. Endothelial destabilization by angiopoietin-2 via integrin β1 activation. Nat Commun. 2015;6:5962.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 165]  [Cited by in RCA: 215]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
40.  Hakanpaa L, Kiss EA, Jacquemet G, Miinalainen I, Lerche M, Guzmán C, Mervaala E, Eklund L, Ivaska J, Saharinen P. Targeting β1-integrin inhibits vascular leakage in endotoxemia. Proc Natl Acad Sci U S A. 2018;115:E6467-E6476.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 53]  [Cited by in RCA: 76]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
41.  Sui A, Zhong Y, Demetriades AM, Lu Q, Cai Y, Gao Y, Zhu Y, Shen X, Xie B. Inhibition of integrin α5β1 ameliorates VEGF-induced retinal neovascularization and leakage by suppressing NLRP3 inflammasome signaling in a mouse model. Graefes Arch Clin Exp Ophthalmol. 2018;256:951-961.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
42.  Grant PJ. Diabetes mellitus as a prothrombotic condition. J Intern Med. 2007;262:157-172.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 266]  [Cited by in RCA: 251]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
43.  Lemkes BA, Hermanides J, Devries JH, Holleman F, Meijers JC, Hoekstra JB. Hyperglycemia: a prothrombotic factor? J Thromb Haemost. 2010;8:1663-1669.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 196]  [Cited by in RCA: 217]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
44.  Hall R, Suarez S, Majumdar M, Lee I, Zacharias N, Gee D, Dua A. Thromboelastography with Platelet Mapping Identifies High Platelet Reactivity is Associated with Obesity, Diabetes, and Thrombotic Events. Ann Vasc Surg. 2024;104:227-236.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
45.  Heath H, Brigden WD, Canever JV, Pollock J, Hunter PR, Kelsey J, Bloom A. Platelet adhesiveness and aggregation in relation to diabetic retinopathy. Diabetologia. 1971;7:308-315.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 148]  [Cited by in RCA: 151]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
46.  Gong H, Song Q, Wang L. Manifestations of central retinal artery occlusion revealed by fundus fluorescein angiography are associated with the degree of visual loss. Exp Ther Med. 2016;11:2420-2424.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
47.  Ju L, McFadyen JD, Al-Daher S, Alwis I, Chen Y, Tønnesen LL, Maiocchi S, Coulter B, Calkin AC, Felner EI, Cohen N, Yuan Y, Schoenwaelder SM, Cooper ME, Zhu C, Jackson SP. Compression force sensing regulates integrin α(IIb)β(3) adhesive function on diabetic platelets. Nat Commun. 2018;9:1087.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 44]  [Cited by in RCA: 40]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
48.  Takada YK, Fujita M, Takada Y. Pro-Inflammatory Chemokines CCL5, CXCL12, and CX3CL1 Bind to and Activate Platelet Integrin αIIbβ3 in an Allosteric Manner. Cells. 2022;11:3059.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
49.  Niu H, Chen X, Gruppo RA, Li D, Wang Y, Zhang L, Wang K, Chai W, Sun Y, Ding Z, Gartner TK, Liu J. Integrin αIIb-mediated PI3K/Akt activation in platelets. PLoS One. 2012;7:e47356.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
50.  Qin RR, Zhu H, Wang F, Song M, Lin PL, Xing YQ, Zhang W, Zhong M, Wang ZH. Platelet activation in diabetic mice models: the role of vascular endothelial cell-derived protein disulfide isomerase-mediated GP IIb/IIIa receptor activation. Aging (Albany NY). 2019;11:6358-6370.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
51.  Hood JD, Frausto R, Kiosses WB, Schwartz MA, Cheresh DA. Differential alphav integrin-mediated Ras-ERK signaling during two pathways of angiogenesis. J Cell Biol. 2003;162:933-943.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 203]  [Cited by in RCA: 202]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
52.  Eliceiri BP, Puente XS, Hood JD, Stupack DG, Schlaepfer DD, Huang XZ, Sheppard D, Cheresh DA. Src-mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. J Cell Biol. 2002;157:149-160.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 265]  [Cited by in RCA: 271]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
53.  Mahabeleshwar GH, Feng W, Reddy K, Plow EF, Byzova TV. Mechanisms of integrin-vascular endothelial growth factor receptor cross-activation in angiogenesis. Circ Res. 2007;101:570-580.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 225]  [Cited by in RCA: 238]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
54.  Drake CJ, Cheresh DA, Little CD. An antagonist of integrin alpha v beta 3 prevents maturation of blood vessels during embryonic neovascularization. J Cell Sci. 1995;108 ( Pt 7):2655-2661.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 227]  [Cited by in RCA: 230]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
55.  Scheppke L, Murphy EA, Zarpellon A, Hofmann JJ, Merkulova A, Shields DJ, Weis SM, Byzova TV, Ruggeri ZM, Iruela-Arispe ML, Cheresh DA. Notch promotes vascular maturation by inducing integrin-mediated smooth muscle cell adhesion to the endothelial basement membrane. Blood. 2012;119:2149-2158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 99]  [Cited by in RCA: 112]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
56.  Nebbioso M, Lambiase A, Cerini A, Limoli PG, La Cava M, Greco A. Therapeutic Approaches with Intravitreal Injections in Geographic Atrophy Secondary to Age-Related Macular Degeneration: Current Drugs and Potential Molecules. Int J Mol Sci. 2019;20:1693.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 33]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
57.  Karageozian V  Phase 2 Randomized Clinical Trial of Luminate® as Compared to Avastin® in the Treatment of Diabetic Macular Edema. [accessed 2024 Dec 19]. In: ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT02348918 ClinicalTrials.gov Identifier: NCT02348918.  [PubMed]  [DOI]
58.  Khanani AM, Patel SS, Gonzalez VH, Moon SJ, Jaffe GJ, Wells JA, Kozma P, Dugel PU, Maturi RK. Phase 1 Study of THR-687, a Novel, Highly Potent Integrin Antagonist for the Treatment of Diabetic Macular Edema. Ophthalmol Sci. 2021;1:100040.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
59.  Oxurion  A Study to Evaluate the Safety of THR-687 in Subjects With Diabetic Macular Edema (DME). [accessed 2024 Dec 19]. In: ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT03666923 ClinicalTrials.gov Identifier: NCT03666923.  [PubMed]  [DOI]
60.  Oxurion  A Study to Evaluate THR-687 Treatment for Diabetic Macular Oedema. [accessed 2024 Dec 19]. In: ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT05063734 ClinicalTrials.gov Identifier: NCT05063734.  [PubMed]  [DOI]
61.  Askew BC, Furuya T, Edwards DS. Ocular Distribution and Pharmacodynamics of SF0166, a Topically Administered α(v)β(3) Integrin Antagonist, for the Treatment of Retinal Diseases. J Pharmacol Exp Ther. 2018;366:244-250.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
62.  Foulks G  Safety and Exploratory Efficacy Study of SF0166 for the Treatment of Diabetic Macular Edema (DME). [accessed 2024 Dec 19]. In: ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT02914613 ClinicalTrials.gov Identifier: NCT02914613.  [PubMed]  [DOI]
63.  Regillo C  Nesvategrast (OTT166) in Diabetic Retinopathy (DR). [accessed 2024 Dec 19]. In: ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT05409235 ClinicalTrials.gov Identifier: NCT05409235.  [PubMed]  [DOI]
64.  Yun SH, Sim EH, Goh RY, Park JI, Han JY. Platelet Activation: The Mechanisms and Potential Biomarkers. Biomed Res Int. 2016;2016:9060143.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 294]  [Cited by in RCA: 316]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
65.  O'Shea JC, Hafley GE, Greenberg S, Hasselblad V, Lorenz TJ, Kitt MM, Strony J, Tcheng JE; ESPRIT Investigators (Enhanced Suppression of the Platelet IIb/IIIa Receptor with Integrilin Therapy trial). Platelet glycoprotein IIb/IIIa integrin blockade with eptifibatide in coronary stent intervention: the ESPRIT trial: a randomized controlled trial. JAMA. 2001;285:2468-2473.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 165]  [Cited by in RCA: 160]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
66.  O'Shea JC, Buller CE, Cantor WJ, Chandler AB, Cohen EA, Cohen DJ, Gilchrist IC, Kleiman NS, Labinaz M, Madan M, Hafley GE, Califf RM, Kitt MM, Strony J, Tcheng JE; ESPRIT Investigators. Long-term efficacy of platelet glycoprotein IIb/IIIa integrin blockade with eptifibatide in coronary stent intervention. JAMA. 2002;287:618-621.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 75]  [Cited by in RCA: 78]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
67.  Piccolo R, Eitel I, Galasso G, Iversen AZ, Gu YL, Dominguez-Rodriguez A, de Smet BJ, Mahmoud KD, Abreu-Gonzalez P, Thiele H, Piscione F. Intracoronary abciximab in diabetic patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. Vascul Pharmacol. 2015;73:32-37.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
68.  Piccolo R, Eitel I, Galasso G, Dominguez-Rodriguez A, Iversen AZ, Abreu-Gonzalez P, Windecker S, Thiele H, Piscione F. 1-Year Outcomes With Intracoronary Abciximab in Diabetic Patients Undergoing Primary Percutaneous Coronary Intervention. J Am Coll Cardiol. 2016;68:727-738.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
69.  Chew DP, Bhatt DL, Sapp S, Topol EJ. Increased mortality with oral platelet glycoprotein IIb/IIIa antagonists: a meta-analysis of phase III multicenter randomized trials. Circulation. 2001;103:201-206.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 253]  [Cited by in RCA: 229]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
70.  Ghosh N, Garg I, Srivastava S, Kumar B. Influence of integrins on thrombus formation: a road leading to the unravelling of DVT. Mol Cell Biochem. 2021;476:1489-1504.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
71.  Vootukuri S, Li J, Nedelman M, Thomas C, Jiang JK, Babayeva M, Coller BS. Preclinical Studies of RUC-4, a Novel Platelet αIIbβ3 Antagonist, in Non-Human Primates and With Human Platelets. J Clin Transl Sci. 2019;3:65-74.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
72.  Kereiakes DJ, Henry TD, DeMaria AN, Bentur O, Carlson M, Seng Yue C, Martin LH, Midkiff J, Mueller M, Meek T, Garza D, Gibson CM, Coller BS. First Human Use of RUC-4: A Nonactivating Second-Generation Small-Molecule Platelet Glycoprotein IIb/IIIa (Integrin αIIbβ3) Inhibitor Designed for Subcutaneous Point-of-Care Treatment of ST-Segment-Elevation Myocardial Infarction. J Am Heart Assoc. 2020;9:e016552.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 20]  [Cited by in RCA: 20]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
73.  Shojaei A  Safety and Bioactivity of AXT107 in Subjects With Diabetic Macular Edema (CONGO). [accessed 2024 Dec 19]. In: ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT04697758 ClinicalTrials.gov Identifier: NCT04697758.  [PubMed]  [DOI]
74.  Bhatwadekar AD, Kansara V, Luo Q, Ciulla T. Anti-integrin therapy for retinovascular diseases. Expert Opin Investig Drugs. 2020;29:935-945.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
75.  Boyer DS, Kaiser PK, Magrath GN, Brady K, Edwards S, Tanzer DJ, Heier JS. The Safety and Biological Activity of OTT166, a Novel Topical Selective Integrin Inhibitor for the Treatment of Diabetic Eye Disease: A Phase 1b Study. Ophthalmic Surg Lasers Imaging Retina. 2022;53:553-560.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
76.  Diagram BV  A Phase 2 Open Label Study to Assess the PK/PD Properties of RUC-4 in Patients With a ST-elevation Myocardial Infarction (CEL-02). [accessed 2024 Dec 19]. In: ClinicalTrials.gov [Internet]. Bethesda (MD): U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT04284995 ClinicalTrials.gov Identifier: NCT04284995.  [PubMed]  [DOI]