Downloads

Meredith, E., & A. Schwartz, M. Integrins as Drug Targets in Vascular and Related Diseases. International Journal of Drug Discovery and Pharmacology. 2024, 3(2), 100010. doi: https://doi.org/10.53941/ijddp.2024.100010

Review

Integrins as Drug Targets in Vascular and Related Diseases

Emily Meredith 1,*, and Martin A. Schwartz 1,2,3

1  Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511, USA, martin.schwartz@yale.edu

2  Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520, USA.

3  Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA.

* Correspondence: Emily.meredith@yale.edu

Received: 6 April 2024; Revised: 28 May 2024; Accepted: 29 May 2024; Published: 21 June 2024

Abstract: Integrins are transmembrane receptors that, as critical participants in a vast range of pathological processes, are potential therapeutic targets. However, in only a few cases has the promise been realized by drug approval. In this review, we briefly review basic integrin biology and participation in disease, challenges in the development of safe, effective integrin-targeted therapies, and recent advances that may lead to progress.

Keywords:

Integrins cardiovascular disease integrin therapeutics

1. Introduction

Integrins are heterodimeric transmembrane receptors that bind extracellular matrix (ECM) proteins and counter receptors on other cells, thereby mediating cell migration, angiogenesis, inflammatory signaling and immune cell invasion [1]. There are 24 unique integrin dimers, each formed by pairing one of the 18 alpha (α) subunits with one of the 8 beta (β) subunits, generating receptors with distinct ligand-binding and signaling functions. Integrins connect to the actin cytoskeleton inside the cell through a network of linker proteins, among which talin is the most crucial. This linkage transmits force between the ECM and the cytoskeleton, conferring mechanical stability to cells and tissues.

In addition to binding ECM components such as fibronectin, vitronectin, collagen and laminin, integrins transmit signals to the cytoplasm through multiple adapters and signaling proteins, so-called outside-in signaling [2]. Conversely, binding of cytoplasmic proteins to integrin cytoplasmic tails can trigger a change in integrin conformation leading to increased affinity for extracellular ligands, termed inside-out signaling or activation. Integrin bidirectional signaling is highly dependent on complex conformational transitions that have been refined over 20 years of structural studies. In brief, bent, low-affinity integrins move through a series of steps to open, extended high affinity conformations; these conformational transitions are also dependent on mechanical loads such that tension promotes or stabilizes the extended, high affinity states [3]. In this way, integrins enable cells to both exert and sense ECM mechanical properties such as stiffness, loading and topography. Integrins transduce structural and mechanical variables into biochemical signals that guide a vast range of biological processes.

Reagents targeting GPCRs make up a staggering 40% of marketed drugs as of 2022 [4]. Here, specificity is enabled by the 850 GPCRs in the human genome and the extensive structural biology studies that have led to development of high affinity antagonists and agonists. New tools such as AlphaFold2, are further accelerating progress [5]. With only 24 integrins that often share subunits, development of successful integrin-targeting drugs is more challenging. Progress thus requires deeper understanding of both integrin structural biology and functional biology in physiological and pathological processes.

While this review focuses on vascular and related diseases, we define these terms broadly, in keeping with the centrality of the vasculature to the physiology and pathology of nearly every other organ and system. Blood vessels control the growth and spread of cancers, the trafficking of leukocytes in inflammatory diseases, and health of many organs apart from basic functions of blood transport, hemostasis and repair. Thus, we will briefly review basic integrin expression, function and structural biology before covering contrasting successful and unsuccessful integrin therapies, where unsuccessful therapies fell short, and how we can move toward developing safe and effective integrin-targeted therapies.

2. Integrins in Disease

Figure 1. Regulation of integrins in vascular and related diseases. Atherosclerosis, aneurysms, pulmonary arterial hypertension (PAH), fibrosis, thrombosis and cancer involve multiple cell types in which altered integrin expression and/or function contribute to pathology. Endothelial cells (ECs), vascular smooth muscle cells (VSMCs), leukocytes, fibroblasts and platelets all change their integrin expression and activation profile in these disease states, α5β1 and αv integrins are upregulated and or activated in many of these settings, in keeping with their general roles in cell growth and migration during dynamic processes. The leukocyte integrins (β2 and α4 families) also contribute to inflammatory diseases via effects on leukocyte migration, activation and effector function. These integrins have this been the main targets for translational and clinical studies.

Vascular diseases in which integrins are central include atherosclerosis, thrombosis, pulmonary arterial hypertension (PAH) and thoracic and abdominal aortic aneurysms (TAA, AAA, respectively). The vasculature and integrins expressed therein also play supporting but important roles in other conditions, such as cancer, autoimmune disorders and fibrosis.

The endothelial cells that line the blood vessels control permeability and movement of leukocytes, mediate angiogenesis, control vascular tone and generally orchestrate blood vessel growth and function [6]. Fluid shear stress from blood flow exerts a major regulatory influence on endothelial phenotype, a process to which integrins substantially contribute [7]. Atherosclerosis arises selectively in regions of arteries under disturbed flow patterns (DSS). These regions are found at branch points and sites of high curvature, where changes in endothelial phenotype are associated with changes in integrin expression and ECM remodeling [8,9]. Both endothelial and smooth muscle compartments show increased expression and assembly of a fibronectin matrix in atherosclerosis, increased expression of fibronectin-binding integrins αvβ3 and α5β1, together with integrin activation, which together enhance inflammatory activation and plaque progression [10]. Importantly, inflamed endothelial cells express and activate Transforming Growth Factor-beta (TGFβ) signaling and undergo endothelial-mesenchymal transition (EndMT) under these conditions. Fibronectin and integrin α5 are both Smad2/3 target genes that are upregulated during EndMT [11] and blocking fibronectin-integrin signaling potently reduced atherosclerosis in mice [12,‍13]. The FN-integrin interaction and downstream signaling thus appears to be an important circuit within the TGFβ/EndMT/inflammatory cascade. Vascular smooth muscle cells (VSMCs) in the plaque also increase their expression of αvβ3 and α5β1 integrins, and decrease integrins associated with the contractile state, promoting phenotypic switching, migration into and proliferation in the plaque and TGFβ activation [14], creating an additional feed forward mechanism that amplifies disease [15,16].

Infiltrating immune cells are equally important to the above diseases. T cells, macrophages and neutrophils are vital in tissue surveillance to detect damage and infection. This process requires their exit from the blood stream and subsequent migration through the tissue via integrins such as LFA-1 (αLβ2), Mac-1 (αMβ2), αxβ2, and α4β1 [17]. The migratory processes require integrins that bind their ligands with high affinity, thus, integrin activation is also critical for tissue inflammation, as well as other aspects of immunity. Inflammatory mediators trigger conversion of these integrins to the high affinity state to enable immune cell function [17]. In atherosclerosis, infiltrating monocytes are central to vessel inflammatory signaling. Blocking these processes is a viable approach for combatting pathological inflammation in, for example, autoimmune disease but the dangers of suppressing host pathogen defense must also be considered.

Major risk factors that can lead to the development of atherosclerosis include high blood pressure, elevated circulating cholesterol levels, and obesity. Other arterial diseases, including pulmonary arterial hypertension (PAH) and aneurysms, share both risk factors and pathological processes, and are often diagnosed concurrently or as co-morbidities [1821]. In PAH, lung endothelial cells increase their integrin expression during artery narrowing associated with elevated blood pressure [22]. Endothelial cells upregulate integrin β5, while VSMCs upregulate αvβ3 [22,23]. Leukocytes, mainly activated macrophages and T cells, also infiltrate into remodeled pulmonary arteries of PAH patients associated with integrin activation [24].

A major component in atherosclerosis, hypertensive artery remodeling and cancer is fibrosis, mediated mainly by myofibroblasts. These cells derive from fibroblasts, which involves upregulation of integrin α5β1 as well as α6β1, αvβ6 and α8β1 and downregulating integrins associated with inactive fibroblasts [25,26] Myofibroblasts are the highly contractile and secretory cells that make the dense collagen matrix associated with fibrosis. Fibrosis is of course associated with disease states in many other organs, which involves similar transitions in integrin expression and function [27].

Aneurysms represent a form of pathological remodeling due to some combination of high blood pressure, mutations in ECM, contractile proteins and their regulators, and inflammation [28]. Integrin α5β1 on SMCs was found to be critical to the progression and rupture of aneurysms through the fibronectin-mediated inflammatory signaling cascade [29]. Integrin αvβ3 also makes an important contribution, involving both altered expression [30] and changes in its ability to bind ECM ligands due to induction of legumain in macrophages [31]. Finally, integrin-mediated infiltration of leukocytes, contributes to aneurysm instability and perpetuates the inflammatory environment [28].

In cancer, αvβ3 and α5β1 integrins are often upregulated on both tumor cells and on the endothelial cells that mediate tumor angiogenesis to enable tumor growth and dissemination [3234]. Work targeting vascular integrins as cancer therapy remains unsuccessful due to some combination of low efficacy and side effects [35]. Again, drug development requires balancing integrin roles in these pathological processes with beneficial roles in vascular homeostasis.

Arguably the most straightforward condition in which integrins are a primary target is thrombosis. Blood platelets that mediate clotting are unique in expressing integrin αIIbβ3. Under normal circumstances, platelets integrins are inactive, allowing platelets to circulate freely. Upon vascular injury, activation of the clotting cascade triggers signals within platelets that result in conformational activation of integrin αIIbβ3 to increase its affinity for fibrinogen/fibrin and other ligands, promoting platelet aggregation and thus clotting at the wound site [36]. Platelets also show partial activation in inflammatory environments, which increases their propensity for thrombosis [37]. αIIbβ3 antagonists have thus been developed to inhibit pathological thrombosis, though are used with great care to avoid bleeding complications [38].

Thus, while integrins’ essentiality makes them attractive therapeutic targets, their physiological roles place severe constraints. Precise and proper regulation of hemostasis and immunity is required for health. Additionally, vascular homeostasis is an ongoing process. As tissues grow, undergo changes in metabolism or age, demand for vascularization changes. Enabling the circulation to meet tissue metabolic requirements requires its ongoing adaptation via physiological angiogenesis and arteriogenesis. Inadequate vascularity (vascular rarefaction) in aging is a major cause of heart failure, frailty, and other problems [39]. Precision, cautious and careful evaluation of on-target but unwanted effects is thus required.

2. Current Methods of Targeting Integrins and Their Pathways

Currently marketed therapies are mainly small molecules or monoclonal antibodies (Table 1) that bind integrin extracellular domains to block function by occluding the ligand binding site or by preventing conformational conversion to the high affinity state. The main strategy for the first generation of integrin therapies was to target integrins that were both upregulated in disease and had a relatively limited expression profile. Notable examples include αIIbβ3 that is specific to platelets, α4β1, α4β7 and αLβ2 that are specific to immune cells, and αvβ3 on endothelial cells and tumor cells. However, even with these cell type-limited integrins, complications can arise from on-target but harmful effects in other cell types/tissue locations.

Category Therapy Name Integrin Target Medical Use Clinical Phase Description Ref
Antibody Natalizumab (Tysabri)

α4β1

α4β7

Treating Multiple Sclerosis (MS) FDA approved (2004, 2007), in-use Prevents inflammatory cells from crossing the blood brain barrier and entering the brain, resulting in immunosuppression within the CNS. [40,41]

Abciximab

(ReoPro)

αIIbβ3

αvβ3

Reducing risk of thrombosis during surgery FDA approved (1994), in-use blocks integrins on the surface of platelets, preventing platelet aggregation and reducing the risk of thrombosis. Commonly used in the treatment of acute coronary syndromes. [42]
Agonistic Ligand mimetics 7HP349 α4β1 (VLA-4), αLβ2 (LFA-1)

Vaccine adjuvant, treating

metastatic melanoma

FDA Fast Track Status (2022) Small molecule binding activates VLA-4 and LFA-1 on leukocytes, increasing their adhesiveness to promote VLA-4/VCAM-1- and LFA-1/ICAM-1-mediated adhesion between naive T cells and antigen presenting cells (APCs) and enhance T-cell activation. [35,43]
Adjacent Pathway Letrozole αvβ3 Infertility FDA approved for the treatment of breast cancer (1998), in-use non-steroidal type II aromatase that inhibits CYP19A1, preventing conversion of androgens to estrogen, thereby reducing uterine weight and elevated leuteinizing hormone. While not approved for infertility specifically, it has been used by physicians for decades to boost IVF effectiveness. [44,45]
Closed-stabilizing Conformational modifiers tirofiban αIIbβ3 Thrombosis inhibitor FDA approved (1999), in-use Small molecule inhibitor that inhibits ADP- and collagen-induced platelet aggregation. [46]
eptifibatide αIIbβ3 Thrombosis inhibitor FDA approved (1998), in-use binds to the platelet integrin αIIb/IIIa of human platelets and inhibits platelet aggregation. [47]
Table 1. FDA-approved integrin-targeting therapies.

Use of the α4 monoclonal antibody (mAb) Natalizumab, approved by the FDA in 2004 for treatment of Multiple Sclerosis (MS), illustrates this complexity. Natalizumab was approved after clinical trials demonstrated an unprecedented 63% reduction in relapse [40,41]. However, it was voluntarily pulled from the market only 1 year later following reports of rare but higher occurrence of progressive multifocal leukoencephalopathy (PML), a devastating viral infection of the white matter of the brain that targets oligodendrocytes [40,48]. Patients, however, petitioned to have it restored, leading to its reapproval in 2007, in part because no comparably effective options were available. It was later found that up to 95% of PML cases occur in patients with a pre-disposing immunodeficiency disorders, such as MS, HIV or hematologic malignancies, and was more frequent in patients on previously available immunosuppressants before starting Natalizumab and in patients taking Natalizumab long-term [49,50]. Recent retrospective studies of the Austrian MS Treatment Registry (AMSTR), founded in 2006, followed Natalizumab patients over a 10 year period [50,51]. In one study [51], 15 Natalizumab patients with diagnosed PML were found to have a mortality of 20% compared to 30-50% in non-MS patients and 25.9% in patients with HIV as the cause of immunodeficiency [52]. While survival is increased relative to non-MS patients, the infection also accelerated MS progression. The Expanded Disability Status Scale (EDSS) increased from 3.5 at pre-PML to 6.5 at the last assessment, leading many patients to require mobility assistance [51]. Furthermore, patients recovered from PML were more likely to convert to progressive MS within three years from PML and the EDSS further increased. In fact, this issue is so prevalent in the treatment of MS that in [50] the authors discovered 53.7% of the patients who discontinued Natalizumab treatment cited either fear of PML or PML diagnosis as the reason for stopping treatment. However, while Natalizumab was the first therapy to be linked to PML, it now appears to be a general consequence of immune suppression [49]. This story serves as both a cautionary tale and a source of hope for future integrin therapies since it indicates that the target integrin may still be feasible if risks of PML can be ameliorated, which would benefit all of the at-risk populations [53].

The αIIbβ3 antagonists Eptifibatide and Tirofiban demonstrate progress toward this goal. These are highly potent anti-thrombotic agents [47,54]. Along with ReoPro, their use is limited to IV administration at low doses and in patients without bleeding complications, precautions necessary due to the potential for bleeding [42,46]. Comparative clinical studies between Eptifibatide and Tirofiban yielded mixed results, although Eptifibatide appeared to offer better safety [5557]. To look more closely into their mechanisms of action, crystallography and structural studies were done, which distinguished between agonistic compounds that stabilize open, active states and antagonistic compounds that stabilize closed, low-affinity states, providing insights into their mechanisms of action [58]. Both compounds halt the transition from bent-closed to open integrin states, thus, Tirofiban and Eptifibatide are considered closed-stabilizing agents; the detailed molecular mechanism is reported to be by binding a water molecule and preventing its expulsion from the ligand binding site during activation [58,59]. This effect is attributed to polar motifs within their structures, particularly piperazine or piperidine, which hydrogen-bond with water [58]. The precise location of these motifs is critical to their function, highlighting potential avenues for therapeutic development targeting RGD-containing integrins. However, challenges persist regarding systemic off-target effects and the role of metal ions like Mg2+ and Mn2+ in ligand binding, again necessitating further exploration in integrin therapy development.

Integrin Clinical Trial Number Phase: Status (Estimated Completion) Pathology Intervention Intervention type
α4β7 NCT05611671 Phase 2: Recruiting (2025) IBD, ulcerative colitis MORF-057 Ligand mimetic
NCT05291689 Phase 2: active (2025)      
NCT04064697 Phase 3: Recruiting (2025) Ulcerative Colitis (UC) with tissue CytoMegaloVirus (CMV) vedolizumab Monoclonal antibody
NCT02768532 Phase 4: Terminated (2023) Crohn’s Disease    
αvβ8 NCT04152018 Phase 1: Active (2024) Solid tumors PF-06940434 Ligand mimetic
αIIbβ3 NCT04284995 Phase 2: Completed (2020) STEMI RUC-4 (Zalunfiban) Ligand mimetic
NCT04825743 Phase 3: recruiting (2024)      
α2 NCT05024994 Phase 2: Active (2024) AML, MDS, CMML E7820 mRNA inhibitor
Table 2. Ongoing clinical trials on integrin-modulating therapies.

3. Clinical Trials Using Integrin Therapies

With these recent advances in integrin structural biology, our understanding of the how small molecules modulate integrin function has expanded to the point that this information can be harnessed to modify existing compounds, generate new ones, or screen currently approved compounds for potential integrin-modifying actions. One significant issue for long term use of integrin targeting compounds is the potential to induce neoepitopes that trigger immune recognition and autoimmunity [60], limiting the time that a patient can stay on the medication. Indeed, new small molecule integrin β3 antagonists have been developed that exhibit increased specificity and potency without exposing neoepitopes [61,62]. RUC-1 and its more potent derivatives RUC-2 and -4 inhibit ligand binding, platelet aggregation and in vivo thrombus formation without inducing integrin conformational changes and accompanying neoepitopes. RUC-4 is currently in clinical trials for the treatment of ST-elevation myocardial infarction (STEMI; NCT04825743). Efficient function blocking without the risk of autoimmunity would be a significant step in integrin-therapy safety that would substantially expand their use.

Along these same lines are a class of drugs known as small-molecule protein ligand interface stabilizers (SPLINTS) [63]. Unlike integrins bound to inhibitors that stabilize the closed or open conformations, SPLITS work intercellularly by stabilizing protein-protein interactions (PPIs), limiting neoepitope formation. A novel SPLINT, E7820, is an aromatic sulphonamide that readily crosses the plasma membrane and triggers degradation of integrin α2 mRNA to inhibit tumor angiogenesis [64,65]. It works as a “molecular glue”, stabilizing a complex between an mRNA splicing co-factor, activator of activating protein 1 and oestrogen receptors (CAPERα) with DDB-1 and cullin-4 associated factor 15 (DCAF15). This complex mediates proteasomal degradation of CAPERα. Through a mechanism yet to be elucidated, the inhibition of this splicing factor prevents the translation of α2 mRNA [63].

Another anti-cancer compound, PF-06940434, targeting αvβ8 is also in development. Among the αv integrins, αvβ8 contains an RGD sequence that seems to prefer latent TGFβ complexes in the ECM [66]. Binding of αvβ8 to TGFβ latent complexes triggers force-dependent release of active TGFβ, a potent cytokine with pleiotropic actions in angiogenesis, cancer growth, tissue repair and fibrosis, and immune suppression [43]. TGFβ signaling is altered in many solid tumors to increase tumor growth [66]. However, global inhibition of TGFβ is not feasible due to serious side effects such as widespread immune activation [43]. Targeting this integrin as a means to inhibit TGFβ activation in a limited way in specific settings may therefore be a useful treatment for cancers or other diseases.

Many of the above-mentioned integrin inhibitors were developed as angiogenesis inhibitors. However, integrin-based anti-angiogenesis clinical trials have been unsuccessful [35]. Importantly, these variable or poor outcomes were not necessarily due to poor inhibition of blood vessel growth but to the difficulty of targeting a disease that can evolve to evade therapy. Angiogenesis inhibitors are problematic because they increase tumor hypoxia, which drives tumor evolution toward more aggressive states [67]. However, angiogenesis inhibitors have more recently been paired with other classes of drugs or radiotherapy. This approach was tested with E7820 for treatment of colorectal cancer, combining it with chemotherapy regimen FOLFIRI (NCT01347645, NCT01133990) or cetuximab (NCT00309179). In ongoing cancer clinical trials using E7820, researchers changed gears from solid-tumor cancers (such as colorectal cancer) to blood and bone marrow cancers such as Acute Myeloid Leukemia (AML), Myelodysplastic Syndrome (MDS) and Chronic myelomonocytic leukemia (CMML) (NCT05024994).

Based on the relative success of therapies such as Natalizumab and 7HP349 that target integrins with greater cell-type specific expression, MORF-057 and vedolizumab are being tested for their efficacy in treating Crohn’s Disease, ulcerative colitis and inflammatory bowel disease (IBD) by targeting α4β7, which is expressed at high levels in memory T cells [68]. However, based on potential complications from brain infection as shown with Natalizumab, safety studies should be followed very closely for other inhibitors of this integrin. Therefore, while short-term safety studies may pass these compounds for clinical use, their long-term can reveal detrimental effects that limit their use.

4. Taking Integrins out of the Picture: Targeting Their Pathways

An alternative approach for targeting integrin functions avoids the intricacies of integrin structure by blocking downstream pathways. For example, phosphodiesterase 4D (PDE4D) binds to and is downstream of integrin α5β1 where it makes an important contribution to vascular inflammation and atherosclerosis [13]. Importantly, the PDE family, specifically PDE3, PDE4 and PDE5, has already been successfully targeted for treatment of other diseases such as erectile dysfunction (PDE5; avanafil, sildenafil, tadalafil, vardenafil), Benign prostatic hyperplasia (PDE5; tadalafil), Pulmonary arterial hypertension (PDE5; sildenafil, tadalafil), Psoriatic arthritis (PDE4; apremilast), Psoriasis (PDE4, apremilast), and cardiovascular disease (reviewed in Bondarev 2022) [69]. Inhibitors of PDE4D may therefore have potential in cardiovascular diseases as a means of blocking integrin-dependent pro-inflammatory signaling without affecting the integrins themselves.

There is also interest in the interplay between integrins and G protein-coupled receptors (GPCRs), driven by the prospect of repurposing the large catalogue of approved GPCR therapies as integrin-targeted treatments. This approach is based on recent work in which selectively inhibiting G protein pathways inhibited integrin outside-in signaling while leaving inside-out signaling intact, i.e., blocking signals without blocking cell adhesion [38,70]. These studies demonstrated a direct link between integrin αIIbβ3 activation and Gα13 activation and showed that blocking this interaction reduced thrombosis in animal models [38,70], suggesting a novel approach to thrombotic disorders.

5. Paths forward in Integrin Therapy

The search for better integrin therapies revolves around the search for higher specificity, based mainly on deeper understanding of integrin structural dynamics. Different integrin pairs assume different resting conformations and variable pathways of activation and ligand binding. For example, 8 of the 24 integrins including αvβ3, αvβ5, αvβ6, αvβ8, α5β1, and αIIbβ3 bind RGD peptides, the structure of the RGD-integrin complexes are quite different depending on the receiving MIDAS binding pocket [71] (reviewed in [72]). Furthermore, conformational dynamics for different integrin dimers depend on numerous factors such as the composition of the ECM and extra- and intra-cellular ligands, which vary depending on biological context. In vitro systems replicate these complex contexts relatively poorly. Differences in the ECM, in expression of intracellular adapters and regulators, and cell state can affect responses to integrin targeting drugs [35]. For example, the αvβ3 antagonists Etaracizumab and VPI-2690B [73,‍74] had strong effects in cell culture systems but stalled in phase 2 clinical trials for failing to lead to clinically meaningful reductions in fibrosis in both several kinds of cancers and in kidney disease [73]. Better in vitro systems such as 3D spheroids and engineered tissue models may offer a path to improved drug development [7579].

Potential off-target and on-target but undesirable effects are major limitations. αv integrins, the most commonly targeted family, are upregulated in cancer and inflammation but still have physiological functions. These integrins play a role in homeostatic vascular remodeling and in bone homeostasis, being constitutively expressed in osteoclasts [80]. One approach to these problems currently under intense development is local delivery. This has been done in mice by implanting biosponges soaked in the αv antagonist IDL-2965 to prevent local muscle fibrosis during regeneration [81]. Specific lipid carriers can target vascular endothelium [82] and hepatocytes [83]. But a good deal of improvement will be required before these approaches gain widespread use in patients. The α5β1 antagonist AXT107 was attached to microparticles for slow release after injection into the eye (NCT05859776) [8486]. Drug-eluting stents inserted into blocked arteries are widely used in the clinic for coronary and periphery artery disease [87,88]; integrin-targeting compounds might be locally delivered via this route as well [89]. However, even in the wall of an atherosclerotic artery, the same integrin can trigger opposite responses depending on the cell type. Integrin αvβ3 is proinflammatory in endothelial cells but promotes the beneficial contractile phenotype in smooth muscle cells [16,90,91]. In endothelial and smooth muscle cells, αvβ3 also promotes migration, which contributes to pathological angiogenesis and atherosclerosis, but has beneficial effects in macrophages/monocytes [92].

Finally, targeting the integrin before it reaches the cell surface may provide a new avenue of drug discovery. For instance, PROTAC (Proteolysis-Targeting Chimeras) technology leverages cell-specific E3 ligases to degrade specific proteins (NCT05573555) [93,94]. This approach could be used in atherosclerosis by exploiting endothelial E3 ligases like HECW2 to target integrins within ECs. There are ongoing clinical trials and studies exploring the applications of PROTAC technology. Combining PROTAC with drug-diffusing stents could improve specific delivery to areas and cells within the vasculature. Alternative PROTACs are being explored that target proteins at the plasma membrane (PM). These approaches offer the potential for localized control over protein degradation and signaling, which could modulate the complex processes in specific cells or locations. However, it is critical to note that inhibiting integrin αvβ3 with small molecules or antibodies gave virtually opposite results compared to genetic ablation. Inhibitors substantially slowed angiogenesis, while genetic deletion aggressively promoted it [95,96]. While it is unknown if this phenomenon will repeat with other integrins, it’s important to keep in mind if PROTAC technology continues to progress. Interestingly, these studies also evaluated and validated the potential for targeting integrin cytoplasmic tails by showing that point mutations in the β3 tail inhibited tumor angiogenesis, again essentially opposite from the effect of β3-knockout in mice [9698].

6. Conclusions

The future of integrin-directed therapies lies in improving specificity. Such improvements may be made by targeting pathways of integrin activation to develop inhibitors that avoid neoepitopes, by developing methods to target specific cell types or tissues, or by targeting downstream pathways that show more restricted expression/utilization. The very broad importance of integrins in biological and pathological processes supports their great potential as therapeutic targets, reviewed in [35,38,72]. But the limited number of approved compounds to date underscores the need for a deeper understanding of underlying issues, such as target specificity and conformation. Thus, advances in drug delivery and PROTAC development combined with deeper understanding of integrin structural biology and signaling defines the path forward.

Institutional Review Board Statement: Not applicable.

Informed Consent Statement: Not applicable.

References

  1. Takada, Y.; Ye, X.; Simon, S. The integrins. Genome Biol. 2007, 8, 215. DOI: https://doi.org/10.1186/gb-2007-8-5-215
  2. Hynes, R.O. The emergence of integrins: A personal and historical perspective. Matrix Biol. 2004, 23, 333–340, https://doi.org/10.1016/j.matbio.2004.08.001. DOI: https://doi.org/10.1016/j.matbio.2004.08.001
  3. Morse, E.M.; Brahme, N.N.; Calderwood, D.A. Integrin Cytoplasmic Tail Interactions. Biochemistry 2014, 53, 810–820, https://doi.org/10.1021/bi401596q. DOI: https://doi.org/10.1021/bi401596q
  4. Laeremans, T.; Sands, Z.A.; Claes, P.; et al. Accelerating GPCR drug discovery with conformation-stabilizing VHHs. Front. Mol. Biosci. 2022, 9, 863099, https://doi.org/10.3389/fmolb.2022.863099. DOI: https://doi.org/10.3389/fmolb.2022.863099
  5. Zhang, H.; Zhu, D.S.; Zhu, J. Family-wide analysis of integrin structures predicted by αFold2. Comput. Struct. Biotechnol. J. 2023, 21, 4497–4507, https://doi.org/10.1016/j.csbj.2023.09.022. DOI: https://doi.org/10.1016/j.csbj.2023.09.022
  6. Trimm, E.; Red-Horse, K. Vascular endothelial cell development and diversity. Nat. Rev. Cardiol. 2022, 20, 197–210, https://doi.org/10.1038/s41569-022-00770-1. DOI: https://doi.org/10.1038/s41569-022-00770-1
  7. Baeyens, N.; Bandyopadhyay, C.; Coon, B.G.; et al. Endothelial fluid shear stress sensing in vascular health and disease. J. Clin. Investig. 2016, 126, 821–828, doi:10.1172/jci83083. DOI: https://doi.org/10.1172/JCI83083
  8. Li, F.; Yan, K.; Wu, L.; et al. Single-cell RNA-seq reveals cellular heterogeneity of mouse carotid artery under disturbed flow. Cell. Death Discov. 2021, 7, 1–14, https://doi.org/10.1038/s41420-021-00567-0. DOI: https://doi.org/10.1038/s41420-021-00567-0
  9. Tzima, E.; del Pozo, M.A.; Shattil, S.J.; et al. Activation of integrins in endothelial cells by fluid shear stress mediates Rho-dependent cytoskeletal alignment. EMBO J. 2001, 20, 4639–4647, https://doi.org/10.1093/emboj/20.17.4639. DOI: https://doi.org/10.1093/emboj/20.17.4639
  10. Finney, A.C.; Stokes, K.Y.; Pattillo, C.B.; et al. Integrin signaling in atherosclerosis. Cell. Mol. Life Sci. 2017, 74, 2263–2282, https://doi.org/10.1007/s00018-017-2490-4. DOI: https://doi.org/10.1007/s00018-017-2490-4
  11. Chen, P.Y.; Qin, L.; Baeyens, N.; et al. Endothelial-to-mesenchymal transition drives atherosclerosis progression. J. Clin. Invest. 2015, 125, 4514–4528. DOI: https://doi.org/10.1172/JCI82719
  12. Yun, S.; Budatha, M.; Dahlman, J.E.; et al. Interaction between integrin α5 and PDE4D regulates endothelial inflammatory signalling. Nature 2016, 18, 1043–1053, https://doi.org/10.1038/ncb3405. DOI: https://doi.org/10.1038/ncb3405
  13. Yun, S.; Hu, R.; Schwaemmle, M.E.; et al. Integrin α5β1 regulates PP2A complex assembly through PDE4D in atherosclerosis. J. Clin. Invest. 2019, 129, 4863–4874. DOI: https://doi.org/10.1172/JCI127692
  14. Piera-Velazquez, S.; Jimenez, S.A. Endothelial to mesenchymal transition: Role in physiology and in the pathogenesis of human diseases. Physiol. Rev. 2019, 99, 1281–1324, https://doi.org/10.1152/physrev.00021.2018. DOI: https://doi.org/10.1152/physrev.00021.2018
  15. Allahverdian, S.; Chaabane, C.; Boukais, K.; et al. Smooth muscle cell fate and plasticity in atherosclerosis. Cardiovasc. Res. 2018, 114, 540–550, doi:10.1093/cvr/cvy022. DOI: https://doi.org/10.1093/cvr/cvy022
  16. Misra, A.; Feng, Z.; Chandran, R.R.; et al. Integrin beta3 regulates clonality and fate of smooth muscle-derived atherosclerotic plaque cells. Nat. Commun. 2018, 9, 2073, https://doi.org/10.1038/s41467-018-04447-7. DOI: https://doi.org/10.1038/s41467-018-04447-7
  17. Zhang, Y.; Wang, H. Integrin signalling and function in immune cells. Immunology 2012, 135, 268–275, https://doi.org/10.1111/j.1365-2567.2011.03549.x. DOI: https://doi.org/10.1111/j.1365-2567.2011.03549.x
  18. Renshall, L.; Arnold, N.; West, L.; et al. Selective improvement of pulmonary arterial hypertension with a dual ET(A)/ET(B) receptors antagonist in the apolipoprotein E-/- model of PAH and atherosclerosis. Pulm. Circ. 2018, 8, 2045893217752328. DOI: https://doi.org/10.1177/2045893217752328
  19. Reed, D.; Reed, C.; Stemmermann, G.; et al. Are aortic aneurysms caused by atherosclerosis? Circulation 1992, 85, 205–211. DOI: https://doi.org/10.1161/01.CIR.85.1.205
  20. Nickel, N.P.; Yuan, K.; Dorfmuller, P.; et al., Beyond the lungs: Systemic manifestations of pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 2020, 201, 148–157. DOI: https://doi.org/10.1164/rccm.201903-0656CI
  21. Maleki, S.; Poujade, F.-A.; Bergman, O.; et al. Endothelial/epithelial mesenchymal transition in ascending aortas of patients with bicuspid aortic valve. Front. Cardiovasc. Med. 2019, 6, 182, https://doi.org/10.3389/fcvm.2019.00182. DOI: https://doi.org/10.3389/fcvm.2019.00182
  22. Jia, D.; Zhu, Q.; Liu, H.; et al. Osteoprotegerin disruption attenuates hysu-induced pulmonary hypertension through integrin αvβ3/FAK/AKT pathway suppression. Circ. Cardiovasc Genet. 2017, 10, e001591. DOI: https://doi.org/10.1161/CIRCGENETICS.116.001591
  23. Blanchard, N.; Link, P.A.; Farkas, D.; et al., Dichotomous role of integrin-β5 in lung endothelial cells. Pulm. Circ. 2022, 12, e12156. DOI: https://doi.org/10.1002/pul2.12156
  24. Andre, P.; Joshi, S.R.; Briscoe, S.D.; et al. Therapeutic approaches for treating pulmonary arterial hypertension by correcting imbalanced TGF-β superfamily signaling. Front. Med. 2021, 8, 814222. DOI: https://doi.org/10.3389/fmed.2021.814222
  25. Shochet, G.E.; Brook, E.; Bardenstein-Wald, B.; et al. Integrin α-5 silencing leads to myofibroblastic differentiation in IPF-derived human lung fibroblasts. Ther. Adv. Chronic Dis. 2020, 11, 2040622320936023, https://doi.org/10.1177/2040622320936023. DOI: https://doi.org/10.1177/2040622320936023
  26. Chen, H.; Qu, J.; Huang, X.; et al. Mechanosensing by the α6-integrin confers an invasive fibroblast phenotype and mediates lung fibrosis. Nat. Commun. 2016, 7, 12564, https://doi.org/10.1038/ncomms12564. DOI: https://doi.org/10.1038/ncomms12564
  27. Manso, A.M.; Kang, S.M.; Ross, R.S. Integrins, focal adhesions, and cardiac fibroblasts. J. Investig. Med. 2009, 57, 856–60. DOI: https://doi.org/10.2310/JIM.0b013e3181c5e61f
  28. Guo, D.C.; Papke, C.L.; He, R.; et al. Pathogenesis of thoracic and abdominal aortic aneurysms. Ann. N.Y. Acad. Sci. 2006, 1085, 339–352. DOI: https://doi.org/10.1196/annals.1383.013
  29. Chen, M.; Cavinato, C.; Hansen, J.; et al. FN (Fibronectin)-Integrin α5 signaling promotes thoracic aortic aneurysm in a mouse model of marfan syndrome. Arter. Thromb. Vasc. Biol. 2023, 43, E132–E150, https://doi.org/10.1161/atvbaha.123.319120. DOI: https://doi.org/10.1161/ATVBAHA.123.319120
  30. Nakamura, K.; Dalal, A.R.; Yokoyama, N.; et al. Lineage-Specific induced pluripotent stem cell–derived smooth muscle cell modeling predicts integrin alpha-v antagonism reduces aortic root aneurysm formation in marfan syndrome mice. Arter. Thromb. Vasc. Biol. 2023, 43, 1134–1153, https://doi.org/10.1161/atvbaha.122.318448. DOI: https://doi.org/10.1161/ATVBAHA.122.318448
  31. Pan, L.; Bai, P.; Weng, X.; et al., Legumain is an endogenous modulator of integrin αvβ3 triggering vascular degeneration, dissection, and rupture. Circulation 2022, 145, 659–674. DOI: https://doi.org/10.1161/CIRCULATIONAHA.121.056640
  32. Wu, P.H.; Opadele, A.E.; Onodera, Y.; et al., Targeting integrins in cancer nanomedicine: Applications in cancer diagnosis and therapy. Cancers 2019, 11, 1783. DOI: https://doi.org/10.3390/cancers11111783
  33. Hamidi, H.; Ivaska, J. Every step of the way: Integrins in cancer progression and metastasis. Nat. Rev. Cancer 2018, 18, 533–548, https://doi.org/10.1038/s41568-018-0038-z. DOI: https://doi.org/10.1038/s41568-018-0038-z
  34. Sloan, E.K.; Pouliot, N.; Stanley, K.L.; et al., Tumor-specific expression of αvβ3 integrin promotes spontaneous metastasis of breast cancer to bone. Breast Cancer Res. 2006, 8, 1–14. DOI: https://doi.org/10.1186/bcr1398
  35. Bergonzini, C.; Kroese, K.; Zweemer, A.J.M.; et al. Targeting integrins for cancer therapy - disappointments and opportunities. Front. Cell. Dev. Biol. 2022, 10, 863850, https://doi.org/10.3389/fcell.2022.863850. DOI: https://doi.org/10.3389/fcell.2022.863850
  36. Janus-Bell, E.; Mangin, P.H. The relative importance of platelet integrins in hemostasis, thrombosis and beyond. Haematologica 2023, 108, 1734–1747, https://doi.org/10.3324/haematol.2022.282136. DOI: https://doi.org/10.3324/haematol.2022.282136
  37. Stark, K.; Massberg, S. Interplay between inflammation and thrombosis in cardiovascular pathology. Nat. Rev. Cardiol. 2021, 18, 666–682, https://doi.org/10.1038/s41569-021-00552-1. DOI: https://doi.org/10.1038/s41569-021-00552-1
  38. Estevez, B.; Shen, B.; Du, X.; et al. Targeting integrin and integrin signaling in treating thrombosis. Arter. Thromb. Vasc. Biol. 2015, 35, 24–29, https://doi.org/10.1161/atvbaha.114.303411. DOI: https://doi.org/10.1161/ATVBAHA.114.303411
  39. Goligorsky, M.S. Microvascular rarefaction: The decline and fall of blood vessels. Organogenesis 2010, 6, 1–10. DOI: https://doi.org/10.4161/org.6.1.10427
  40. Honce, J.M.; Nagae, L.; Nyberg, E. Neuroimaging of natalizumab complications in multiple sclerosis: PML and other associated entities. Mult. Scler. Int. 2015, 2015, 1–14, https://doi.org/10.1155/2015/809252. DOI: https://doi.org/10.1155/2015/809252
  41. Kawamoto, E.; Nakahashi, S.; Okamoto, T.; et al. Anti-integrin therapy for multiple sclerosis. Autoimmune Dis. 2012, 2012, 1–6, https://doi.org/10.1155/2012/357101. DOI: https://doi.org/10.1155/2012/357101
  42. Cohen, S.A.; Trikha, M.; Mascelli, M.A. Potential future clinical applications for the GPIIb/IIIa antagonist, abciximab in thrombosis, vascular and oncological indications. Pathol. Oncol. Res. 2000, 6, 163–174, https://doi.org/10.1007/bf03032368. DOI: https://doi.org/10.1007/BF03032368
  43. Nishimura, S.L. Integrin-mediated transforming growth factor-beta activation, a potential therapeutic target in fibrogenic disorders. Am. J. Pathol. 2009, 175, 1362–1370. DOI: https://doi.org/10.2353/ajpath.2009.090393
  44. Miller, P.B.; Parnell, B.A.; Bushnell, G.; et al. Endometrial receptivity defects during IVF cycles with and without letrozole. Hum. Reprod. 2012, 27, 881–888. DOI: https://doi.org/10.1093/humrep/der452
  45. Chen, Y.F.; Yang, Y.N.; Chu, H.R.; et al. Role of Integrin alphavbeta3 in Doxycycline-Induced Anti-Proliferation in Breast Cancer Cells. Front. Cell Dev. Biol. 2022, 10, 829788. DOI: https://doi.org/10.3389/fcell.2022.829788
  46. Kondo, K.; Umemura, K. Clinical pharmacokinetics of tirofiban, a nonpeptide glycoprotein IIb/IIIa receptor antagonist: Comparison with the monoclonal antibody abciximab. Clin. Pharmacokinet. 2002, 41, 187–195. DOI: https://doi.org/10.2165/00003088-200241030-00003
  47. Tonin, G.; Klen, J. Eptifibatide, an older therapeutic peptide with new indications: From clinical pharmacology to everyday clinical practice. Int. J. Mol. Sci. 2023, 24, 5446, https://doi.org/10.3390/ijms24065446. DOI: https://doi.org/10.3390/ijms24065446
  48. Stuve, O.; Bennett, J.L. Pharmacological properties, toxicology and scientific rationale for the use of natalizumab (Tysabri) in inflammatory diseases. CNS Drug Rev. 2007, 13, 79–95. DOI: https://doi.org/10.1111/j.1527-3458.2007.00003.x
  49. Zaheer, F.; Berger, J.R. Treatment-related progressive multifocal leukoencephalopathy: Current understanding and future steps. Ther. Adv. Drug Saf. 2012, 3, 227–239, https://doi.org/10.1177/2042098612453849. DOI: https://doi.org/10.1177/2042098612453849
  50. Monschein, T.; Dekany, S.; Zrzavy, T.; et al. Real-world use of natalizumab in Austria: Data from the Austrian Multiple Sclerosis Treatment Registry (AMSTR). J. Neurol. 2023, 270, 3779–3786. DOI: https://doi.org/10.1007/s00415-023-11686-2
  51. Moser, T.; Zimmermann, G.; Baumgartner, A.; et al., Long-term outcome of natalizumab-associated progressive multifocal leukoencephalopathy in Austria: A nationwide retrospective study. J. Neurol. 2024, 271, 374–385. DOI: https://doi.org/10.1007/s00415-023-11924-7
  52. Kim, J.; Kim, C.; Lee, J.A.; et al. Long-term prognosis and overall mortality in patients with progressive multifocal leukoencephalopathy. Sci. Rep. 2023, 13, 14291. DOI: https://doi.org/10.1038/s41598-023-41147-9
  53. Anton, R.; Haas, M.; Arlett, P.; et al. Drug-induced progressive multifocal leukoencephalopathy in multiple sclerosis: European regulators’ perspective. Clin. Pharmacol. Ther. 2017, 102, 283–289. DOI: https://doi.org/10.1002/cpt.604
  54. McClellan, K.J.; Goa, K.L. Tirofiban. A review of its use in acute coronary syndromes. Drugs 1998, 56, 1067–1080. DOI: https://doi.org/10.2165/00003495-199856060-00017
  55. Zhou, X.; Wu, X.; Sun, H.; et al. Efficacy and safety of eptifibatide versus tirofiban in acute coronary syndrome patients: A systematic review and meta-analysis. J. Evid. Based Med. 2017, 10, 136–144. DOI: https://doi.org/10.1111/jebm.12253
  56. Puri, A.; Bansal, A.; Narain, V.; et al. Comparative assessment of platelet GpIIb/IIIa receptor occupancy ratio with Eptifibatide/Tirofiban in patients presenting with ACS and undergoing PCI. Indian Hear. J. 2012, 65, 152–157, https://doi.org/10.1016/j.ihj.2012.08.007. DOI: https://doi.org/10.1016/j.ihj.2012.08.007
  57. Movva, H.; Rabah, R.; Tekle, W.; et al. There is no difference in safety and efficacy with Tirofiban or Eptifibatide for patients undergoing treatment of large vessel occlusion and underlying intracranial atherosclerosis. Interdiscip. Neurosurg. 2020, 23, 100927, https://doi.org/10.1016/j.inat.2020.100927. DOI: https://doi.org/10.1016/j.inat.2020.100927
  58. Lin, F.Y.; Li, J.; Xie, Y.; et al., A general chemical principle for creating closure-stabilizing integrin inhibitors. Cell. 2022, 185, 3533–3550. DOI: https://doi.org/10.1016/j.cell.2022.08.008
  59. Zhu, J.; Zhu, J.; Springer, T.A. Complete integrin headpiece opening in eight steps. J. Cell. Biol. 2013. 201, 1053–1068. DOI: https://doi.org/10.1083/jcb.201212037
  60. Abrams, C.S.; Cines, D.B. Thrombocytopenia after treatment with platelet glycoprotein IIb/IIIa inhibitors. Curr. Hemat. Rep. 2004, 3, 143–147.
  61. Blue, R.; Murcia, M.; Karan, C.; et al. Application of high-throughput screening to identify a novel αIIb-specific small- molecule inhibitor of αIIbβ3-mediated platelet interaction with fibrinogen. Blood 2008, 111, 1248–1256. DOI: https://doi.org/10.1182/blood-2007-08-105544
  62. Zhu, J.; Choi, W.S.; McCoy, J.G.; et al. Structure-guided design of a high-affinity platelet integrin αIIbβ3 receptor antagonist that disrupts Mg2+ binding to the MIDAS. Sci. Transl. Med. 2012, 4, 125ra32.
  63. Hülskamp, M.D.; Kronenberg, D.; Stange, R. The small-molecule protein ligand interface stabiliser E7820 induces differential cell line specific responses of integrin α2 expression. BMC Cancer 2021, 21, 1–12, https://doi.org/10.1186/s12885-021-08301-w. DOI: https://doi.org/10.1186/s12885-021-08301-w
  64. Funahashi, Y.; Sugi, N.H.; Semba, T.; et al. Sulfonamide derivative, E7820, is a unique angiogenesis inhibitor suppressing an expression of integrin α2 subunit on endothelium. Cancer Res. 2002, 62, 6116–6123.
  65. Mita, M.; Kelly, K.R.; Mita, A.; et al. Phase I study of E7820, an oral inhibitor of integrin α-2 expression with antiangiogenic properties, in patients with advanced malignancies. Clin. Cancer Res. 2011, 17, 193–200. DOI: https://doi.org/10.1158/1078-0432.CCR-10-0010
  66. McCarty, J.H. αvβ8 integrin adhesion and signaling pathways in development, physiology and disease. J. Cell. Sci. 2020, 133, jcs239434. DOI: https://doi.org/10.1242/jcs.239434
  67. Ansari, M.J.; Bokov, D.; Markov, A.; et al. Cancer combination therapies by angiogenesis inhibitors; a comprehensive review. Cell. Commun. Signal. 2022, 20, 1–23, https://doi.org/10.1186/s12964-022-00838-y. DOI: https://doi.org/10.1186/s12964-022-00838-y
  68. Lokugamage, N.; Chowdhury, I.H.; Biediger, R.J.; et al. Use of a small molecule integrin activator as a systemically administered vaccine adjuvant in controlling Chagas disease. npj Vaccines 2021, 6, 1–14, https://doi.org/10.1038/s41541-021-00378-5. DOI: https://doi.org/10.1038/s41541-021-00378-5
  69. Bondarev, A.D.; Attwood, M.M.; Jonsson, J.; et al. Recent developments of phosphodiesterase inhibitors: Clinical trials, emerging indications and novel molecules. Front. Pharmacol. 2022, 13, 1057083. DOI: https://doi.org/10.3389/fphar.2022.1057083
  70. Shen, B.; Zhao, X.; O’brien, K.A.; et al. A directional switch of integrin signalling and a new anti-thrombotic strategy. Nature 2013, 503, 131–135, https://doi.org/10.1038/nature12613. DOI: https://doi.org/10.1038/nature12613
  71. Kapp, T.G.; Rechenmacher, F.; Neubauer, S.; et al. A comprehensive evaluation of the activity and selectivity profile of ligands for rgd-binding integrins. Sci. Rep. 2017, 7, 39805. DOI: https://doi.org/10.1038/srep39805
  72. Slack, R.J.; Macdonald, S.J.F.; Roper, J.A.; et al. Emerging therapeutic opportunities for integrin inhibitors. Nat. Rev. Drug Discov. 2021, 21, 60–78, https://doi.org/10.1038/s41573-021-00284-4. DOI: https://doi.org/10.1038/s41573-021-00284-4
  73. Hersey, P.; Sosman, J.; O’Day, S.; et al. A randomized phase 2 study of etaracizumab, a monoclonal antibody against integrin αvβ3, + or - dacarbazine in patients with stage IV metastatic melanoma. Cancer 2010, 116, 1526–1534. DOI: https://doi.org/10.1002/cncr.24821
  74. Park, C.H.; Yoo, T.H. TGF-beta inhibitors for therapeutic management of kidney fibrosis. Pharmaceuticals 2022, 15, 1485. DOI: https://doi.org/10.3390/ph15121485
  75. Sant, S.; Johnston, P.A. The production of 3D tumor spheroids for cancer drug discovery. Drug Discov. Today: Technol. 2017, 23, 27–36, https://doi.org/10.1016/j.ddtec.2017.03.002. DOI: https://doi.org/10.1016/j.ddtec.2017.03.002
  76. Ramadan, Q.; Sharma, P. Editorial: Tissue engineering for drug discovery and personalized medicine. Front. Med. Technol. 2021, 3, 663057, https://doi.org/10.3389/fmedt.2021.663057. DOI: https://doi.org/10.3389/fmedt.2021.663057
  77. Jain, P.; Kathuria, H.; Dubey, N. Advances in 3D bioprinting of tissues/organs for regenerative medicine and in-vitro models. Biomaterials 2022, 287, 121639, https://doi.org/10.1016/j.biomaterials.2022.121639. DOI: https://doi.org/10.1016/j.biomaterials.2022.121639
  78. Usman Khan, M.; Cai, X.; Shen, Z.; et al. Challenges in the development and application of organ-on-chips for intranasal drug delivery studies. Pharmaceutics 2023, 15, 1557. DOI: https://doi.org/10.3390/pharmaceutics15051557
  79. Phan, T.H.; Shi, H.; Denes, C.E.; et al. Advanced pathophysiology mimicking lung models for accelerated drug discovery. Biomater. Res. 2023, 27, 35. DOI: https://doi.org/10.1186/s40824-023-00366-x
  80. Zou, W.; Teitelbaum, S.L. Absence of Dap12 and the αvβ3 integrin causes severe osteopetrosis. J. Cell. Biol. 2015, 208, 1251–36. DOI: https://doi.org/10.1083/jcb.201410123
  81. West, C.; Tobo, C.; Au, J.; et al. Combined application of biosponges and an antifibrotic agent for the treatment of volumetric muscle loss. Am. J. Physiol. Physiol. 2023, 324, C1341–C1352, https://doi.org/10.1152/ajpcell.00092.2023. DOI: https://doi.org/10.1152/ajpcell.00092.2023
  82. Dahlman, J.E.; Barnes, C.; Khan, O.F.; et al. In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight. Nat. Nanotechnol. 2014, 9, 648–655, https://doi.org/10.1038/nnano.2014.84. DOI: https://doi.org/10.1038/nnano.2014.84
  83. Cappelluti, M.A.; Poeta, V.M.; Valsoni, S.; et al. Durable and efficient gene silencing in vivo by hit-and-run epigenome editing. Nature 2024, 627, 416–423, https://doi.org/10.1038/s41586-024-07087-8. DOI: https://doi.org/10.1038/s41586-024-07087-8
  84. Lenz, T.; Koch, T.; Joner, M.; et al. Ten‐Year clinical outcomes of biodegradable versus durable polymer new‐generation drug‐eluting stent in patients with coronary artery disease with and without diabetes mellitus. J. Am. Hear. Assoc. 2021, 10, e020165, https://doi.org/10.1161/jaha.120.020165. DOI: https://doi.org/10.1161/JAHA.120.020165
  85. Pandey, N. AXT107 an inhibitor of neovascularization, vascular leakage, and inflammation, is well-tolerated and could potentially be dosed once a year to treat retinal vascular diseases. Invest. Ophthalmology Visual Sci. 2021, 62, 3304–3304.
  86. Joshi, A. Microparticulates for ophthalmic drug delivery. J. Ocul. Pharmacol. Ther. 1994, 10, 29–45, https://doi.org/10.1089/jop.1994.10.29. DOI: https://doi.org/10.1089/jop.1994.10.29
  87. Kim, H.S.; Kang, J.; Hwang, D.; et al. Durable polymer versus biodegradable polymer drug-eluting stents after percutaneous coronary intervention in patients with acute coronary syndrome: The HOST-REDUCE-POLYTECH-ACS trial. Circulation 2021, 143, 1081–1091. DOI: https://doi.org/10.1161/CIRCULATIONAHA.120.051700
  88. Yamaji, K.; Kimura, T.; Morimoto, T.; et al. Very long-term (15 to 20 years) clinical and angiographic outcome after coronary bare metal stent implantation. Circ. Cardiovasc. Interv. 2010, 3, 468–475. DOI: https://doi.org/10.1161/CIRCINTERVENTIONS.110.958249
  89. Blindt, R.; Vogt, F.; Astafieva, I.; et al. A novel drug-eluting stent coated with an integrin-binding cyclic arg-gly-asp peptide inhibits neointimal hyperplasia by recruiting endothelial progenitor cells. J. Am. Coll. Cardiol. 2006, 47, 1786–1795, https://doi.org/10.1016/j.jacc.2005.11.081. DOI: https://doi.org/10.1016/j.jacc.2005.11.081
  90. Wang, W.; Wang, Z.; Tian, D.; et al. Integrin β3 mediates the endothelial-to-mesenchymal transition via the notch pathway. Cell. Physiol. Biochem. 2018, 49, 985. DOI: https://doi.org/10.1159/000493229
  91. Evrard, S.M.; Lecce, L.; Michelis, K.C.; et al. Endothelial to mesenchymal transition is common in atherosclerotic lesions and is associated with plaque instability. Nat. Commun. 2016, 7, 11853, https://doi.org/10.1038/ncomms11853. DOI: https://doi.org/10.1038/ncomms11853
  92. Kabir, I.; Zhang, X.; Dave, J.M.; et al. The age of bone marrow dictates the clonality of smooth muscle-derived cells in atherosclerotic plaques. Nat. Aging 2023, 3, 64–81, https://doi.org/10.1038/s43587-022-00342-5. DOI: https://doi.org/10.1038/s43587-022-00342-5
  93. Bricelj, A.; Steinebach, C.; Kuchta, R.; et al. E3 ligase ligands in successful PROTACs: An overview of syntheses and linker attachment points. Front. Chem. 2021, 9, 707317, https://doi.org/10.3389/fchem.2021.707317. DOI: https://doi.org/10.3389/fchem.2021.707317
  94. Layman, R.M.; Jerzak, K.J.; Hilton, J.F.; et al. TACTIVE-U: Phase 1b/2 umbrella study of ARV-471, a proteolysis targeting chimera (PROTAC) estrogen receptor (ER) degrader, combined with other anticancer treatments in ER+ advanced or metastatic breast cancer. J. Clin. Oncol. 2023, 41, TPS1121–TPS1121, https://doi.org/10.1200/jco.2023.41.16_suppl.tps1121. DOI: https://doi.org/10.1200/JCO.2023.41.16_suppl.TPS1121
  95. Hynes, R.O. A reevaluation of integrins as regulators of angiogenesis. Nat. Med. 2002, 8, 918–921, https://doi.org/10.1038/nm0902-918. DOI: https://doi.org/10.1038/nm0902-918
  96. Demircioglu, F.; Hodivala-Dilke, K. αvβ3 Integrin and tumour blood vessels-learning from the past to shape the future. Curr. Opin. Cell. Biol. 2016, 42, 121–127. DOI: https://doi.org/10.1016/j.ceb.2016.07.008
  97. Mahabeleshwar, G.H.; Feng, W.; Phillips, D.R.; et al. Integrin signaling is critical for pathological angiogenesis. J. Exp. Med. 2006, 203, 2495–2507, https://doi.org/10.1084/jem.20060807. DOI: https://doi.org/10.1084/jem.20060807
  98. Liao, Z.; Kato, H.; Pandey, M.; et al. Interaction of kindlin-2 with integrin β3 promotes outside-in signaling responses by the αVβ3 vitronectin receptor. Blood 2015, 125, 1995–2004. DOI: https://doi.org/10.1182/blood-2014-09-603035