Next Article in Journal
CPR Quality Assessment in Schoolchildren Training
Next Article in Special Issue
Factor XI/XIa Inhibition: The Arsenal in Development for a New Therapeutic Target in Cardio- and Cerebrovascular Disease
Previous Article in Journal
Pre-Stroke Statin Use Is Associated with Mild Neurological Deficits at the Onset of Acute Ischemic Stroke
Previous Article in Special Issue
Left Atrial Appendage Thrombosis and Oral Anticoagulants: A Meta-Analysis of Risk and Treatment Response
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Antithrombotic Agents in Ischemic Cardiovascular Disease: Progress in the Search for the Optimal Treatment

1
Hospital Universitario Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, 25198 Lleida, Spain
2
Institut de Recerca, Hospital Santa Creu i Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
3
Department of Medicine, Autonomous University of Barcelona, 08193 Barcelona, Spain
4
Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CiberCV), 28029 Madrid, Spain
*
Author to whom correspondence should be addressed.
J. Cardiovasc. Dev. Dis. 2022, 9(11), 397; https://doi.org/10.3390/jcdd9110397
Submission received: 3 October 2022 / Revised: 10 November 2022 / Accepted: 11 November 2022 / Published: 16 November 2022

Abstract

:
Ischemic cardiovascular diseases have a high incidence and high mortality worldwide. Therapeutic advances in the last decades have reduced cardiovascular mortality, with antithrombotic therapy being the cornerstone of medical treatment. Yet, currently used antithrombotic agents carry an inherent risk of bleeding associated with adverse cardiovascular outcomes and mortality. Advances in understanding the pathophysiology of thrombus formation have led to the discovery of new targets and the development of new anticoagulants and antiplatelet agents aimed at preventing thrombus stabilization and growth while preserving hemostasis. In the following review, we will comment on the key limitation of the currently used antithrombotic regimes in ischemic heart disease and ischemic stroke and provide an in-depth and state-of-the-art overview of the emerging anticoagulant and antiplatelet agents in the pipeline with the potential to improve clinical outcomes.

1. Introduction

Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. In 2016, 17.9 million people died from all causes of CVDs [1,2]. There were approximately 8.9 million deaths due to ischemic heart disease (IHD) worldwide, remaining the first cause of death; a less prevalent disease was ischemic stroke, with an incidence of 7.6 million globally [3,4,5].
Therapeutic advances in the last decades have reduced CVD mortality, with antithrombotic therapy being the cornerstone of medical treatment. Several antithrombotic drugs are currently used to either block platelet activation (Figure 1), prevent the activation of the coagulation cascade, or induce fibrinolysis once the clot is formed (Figure 2) [6,7,8,9]. Yet, although these antithrombotic agents have robustly demonstrated their effectiveness in preventing atherothrombotic events, they also carry an inherent risk of bleeding. Bleeding is associated with adverse cardiovascular outcomes and mortality; hence, there is a need to discover new targets and develop novel antithrombotic strategies to effectively inhibit thrombosis while preserving hemostasis.
In the following review, we will comment on the key limitations of the currently used antithrombotic regimes in ischemic heart disease and ischemic stroke and provide an in-depth and state-of-the-art overview of the emerging anticoagulant and antiplatelet agents in the pipeline with the potential to improve clinical outcomes.

2. The Coagulation Cascade: Targeting the Intrinsic Coagulation Pathway

Anticoagulants are the treatment of choice to prevent cardioembolic stroke in patients with atrial fibrillation [10,11]. During the last decade, the development of direct oral anticoagulants (DOACs; Figure 2) has brought many advantages as compared to vitamin K antagonists, including a predictable pharmacokinetic profile, rapid onset and offset of action, and fixed dosing with no need for laboratory monitoring or dietary discretion [12]. Conversely, different reversal agents have also been developed to block the effect of anticoagulants in case of need (Table 1).
Yet, important challenges still need to be addressed. As such, bleeding remains the most reported side effect of DOACs, and in certain sub-groups of patients, including patients with mechanical heart valves or triple-positive antiphospholipid disease syndrome, DOACs seem to be less effective than vitamin K antagonists and are not recommended [20,21,22].
Anticoagulants are also implemented in ischemic heart disease since patients who suffer an acute coronary event present an excess of thrombin generation that persists beyond the acute presentation [23]. So far, several trials have demonstrated the ability of anticoagulants to protect against cardiovascular events. As such, the addition of warfarin [24], rivaroxaban [25,26], or ximelagatran [27] to a standard antiplatelet regime has shown to significantly reduce ischemic events, though at the expense of increased bleeding risk. Ximelagatran was, however, withdrawn from the market due to hepatotoxicity and the only anticoagulant recommended by the guidelines for long-term secondary prevention is rivaroxaban, which may be administered at low doses on top of aspirin at 1-year post-MI [25].
Altogether, these trials have evidenced the need to discover new targets that effectively block thrombin generation without displaying hemorrhagic side effects. In recent years, special attention has focused on the main components of the intrinsic coagulation pathway, particularly factor (F)XII, FXI, and FIX [28].

2.1. Targeting Factor XII

FXII has been associated with thrombosis, hereditary angioedema, and (neuro) inflammation. On the other hand, FXII deficiency (i.e., Hageman factor deficiency) is a rare genetic blood disorder that is entirely asymptomatic, showing prolonged active partial thromboplastin times (aPTT) as the only alteration on coagulation tests [29]. FXII circulates in plasma in a zymogen form, and its activation is brought about by the interaction with negatively charged molecules that induce a conformational change in zymogen FXII leading to activated protease FXIIa followed by activation of the enzyme precursors FXI and FIX (Figure 2) [30]. Hence, FXII inhibitors are expected to be particularly efficient in patients whose blood is exposed to non-physiological surfaces of medical devices such as vascular catheters, hemodialysis circuit tubes and membranes, and mechanical valves or stents, that expose negative charge molecules [31]. Alternatively, contact system proteins FXII, high-molecular-weight kininogen (HK), and plasma kallikrein (PK) may assemble on cell surface proteoglycans of various cardiovascular cells. Contact with surface-exposed moieties and plasma-borne soluble contact activators induces FXII activation, which initiates the intrinsic coagulation pathway and activates PK leading to the release of the proinflammatory mediator bradykinin (BK) by PK-mediated cleavage of HK. FXII inhibitors are also being evaluated as a potential treatment for hereditary angioedema, a BK-mediated life-threatening inherited swelling disorder where Serpin C1 esterase inhibitor (a major plasma inhibitor of FXII and and PK) is dysfunctional or deficient [16].
Figure 2. Diagram of the coagulation and fibrinolytic pathways and the different anticoagulants and fibrinolytic agents used in the clinical setting. LMWH: low molecular weight heparin. * Indirect inhibition of factors II, VII, IX and X.
Figure 2. Diagram of the coagulation and fibrinolytic pathways and the different anticoagulants and fibrinolytic agents used in the clinical setting. LMWH: low molecular weight heparin. * Indirect inhibition of factors II, VII, IX and X.
Jcdd 09 00397 g002
Multiple prototypes have been discovered within the last years, including monoclonal antibodies, small interfering RNAs, antisense oligonucleotides, and serine protease inhibitors which are currently being tested at a preclinical level (details are provided in Table 2) [32]. However, each one of these strategies confers different pharmacological properties, which may limit their indications. Antibodies and approaches to silence gene expression require parenteral delivery by subcutaneous or intravenous injection, whereas small molecules can be delivered orally or parenterally (Table 2). On the other hand, small interfering RNAs and antisense oligonucleotides have a slow onset of action requiring about four weeks to achieve therapeutic levels. Although they are not optimal for use in acute settings, their effect extends over time which may enable once-monthly administration. On the other hand, however, they may also require the development of antidotes. In contrast, antibodies and serine protease inhibitors have a rapid onset of action and an expected half-live of <24 h, thereby limiting the need to develop reversal strategies [33].
Garadacimab, a monoclonal antibody, has been the sole FXIIa inhibitor to reach phase III clinical trials (NCT04656418) in patients with hereditary angioedema, showing promising preliminary data after a 6-month follow-up. Another phase III trial (NCT04739059) is ongoing to evaluate its benefits in a longer term (32 months). Based on its proven safety profile and the outcome of both trials, garadacimab may be considered a promising strategy for other indications, including CVDs.

2.2. Targeting Factor XI

Factor XI congenital deficiency has been proven to protect against arterial and venous thrombosis reducing the incidence of deep-vein thrombosis, ischemic stroke, myocardial infarction, and vascular graft occlusion [48,49,50,51]. Most importantly, FXI-deficient patients do not generally exhibit spontaneous bleeding, and the bleeding associated with injury or surgery tends to be mild [52,53]. These observations have supported the development of multiple FXI inhibitors, most of which have reached Phase II testing. Table 3 details the studies conducted so far as per FXI inhibitors.

2.3. Targeting Factor IX

Factor IX is another potential drug target currently under intensive research because of its efficacy and safety profile [72]. Factor IX is activated by both the intrinsic and extrinsic pathways (Figure 2). In the intrinsic pathway, FXIa induces FIX activation, whereas, in the extrinsic coagulation pathway, FIX is activated by the tissue factor (TF)–VIIa complex. FIXa forms a complex with FVIIIa that binds to platelets serving as a very potent activator of FX [73].
As for FXIIa and FXIa, multiple FIX inhibitors have been developed, most of them in the preclinical development phase, and only a few have reached clinical trials (Table 4) [74]. One that raised great interest is pegnivacogin, a RNA-aptamer based FIXa inhibitor featuring a reversal agent, anivamersen [73]. However, both phase II trials where pegnivacogin has been tested have not resulted in the expected positive outcome. The RADAR trial in NSTEMI patients undergoing cardiac catheterization [75] did not show differences between pegnivacogin and heparin, and the REGULATE-PCI trial performed in patients undergoing PCI [76] had to be prematurely terminated due to the presence of severe allergic reactions.

3. Targeting the Platelet: What Is in the Pipeline for Novel Antiplatelet Agents?

Antiplatelet agents are used in treating both ischemic heart disease and ischemic stroke (Figure 1). The currently available antiplatelet agents either: (1) target intraplatelet enzymes (COX-1 inhibition by ASA and PDE inhibition by dipyridamole and cilostazol), preventing the formation of thromboxane A2 (TXA2) or the degradation of AMPc, respectively; or (2) block platelet membrane receptors (P2Y12 receptor antagonists, GPIIb/IIIa inhibitors, and PAR antagonist) preventing their downstream signaling activation (Figure 1) [80].
In the setting of ischemic heart disease, antiplatelet agents are used both in acute and chronic coronary syndromes and after stent implantation to prevent stent-related thrombosis [81,82,83,84]. A double antiplatelet regime with a combination of ASA and a P2Y12 inhibitor is recommended during the first year after an acute myocardial event [85,86,87]. Among the P2Y12 inhibitors, clopidogrel, a second-generation thienopyridine, is a pro-drug that requires a two-enzyme-mediated transformation to become active and irreversibly block the P2Y12 platelet ADP receptors. Yet, clopidogrel exhibits high individual variability because of differences in the activity of cytochrome P450 2C19. Prasugrel, a third-generation thienopyridine, is also a pro-drug but requires fewer hepatic steps to be converted into an active metabolite [88] and hence is less affected by variation in CYP enzymes and exerts a higher degree of platelet inhibition as compared to clopidogrel. Finally, ticagrelor, the first of a new class of P2Y12 inhibitors named cyclopentyl-triazole-pyrimidines, is a reversible P2Y12 receptor inhibitor that does not need hepatic metabolism and accordingly has a more predictable metabolic pathway resulting in a better inter-individual consistency among patients and clinical efficacy. Both prasugrel and ticagrelor have demonstrated greater efficacy than clopidogrel [86,87] and accordingly are recommended over clopidogrel in clinical guidelines in patients with no high bleeding risk [83].
Another known antiplatelet target is the GPIIb/IIIa, the most abundant platelet receptor mainly involved in platelet aggregation [89]. Two GPIIb/IIIa receptor blockers have been approved for intravenous clinical use in STEMI patients, including tirofiban (tyrosine-derived non-peptide derivative) and eptifibatide (heptapeptide). Both antagonize the GPIIb/IIIa receptor preventing fibrinogen and Von Willebrand factor (VWF) from binding to the receptor [90].
In the setting of strokes, antiplatelet therapy is used in secondary prevention in patients with non-cardioembolic transient ischemic attack or stroke. Single antiplatelet therapy with ASA or clopidogrel, or the combination of low dose ASA and dipyridamole or cilostazol, is usually recommended for secondary prevention. In some patients, a combination of ASA and clopidogrel is recommended for up to 90 days to reduce early recurrences [91]. Recent data have demonstrated that ticagrelor on top of ASA reduces the total burden of disability owing to ischemic stroke recurrence compared to ASA alone [92]. Based on these recent findings, the combination of ASA and ticagrelor for up to one month might be considered in patients at risk of ischemic stroke [91]. Cilostazol may also be used for secondary stroke prevention, particularly in Asian patients [93], since randomized clinical trials are still needed to determine its usefulness in non-Asian populations.
There are no reversal agents for the antiplatelet drugs presently used in the clinical setting. However, this might change in short/medium term for ticagrelor. Bentracimab (PB2452) is a recombinant human monoclonal antibody antigen-binding fragment with a dual mechanism of action; it binds both to free ticagrelor and to its major active metabolite (AR-C124910XX) [94]. Bentracimab is currently being tested in a phase III clinical trial (NCT04286438) in patients with uncontrolled major or life-threatening bleeding or requiring urgent survey or invasive procedure.
Despite the currently available antiplatelet armamentarium, recurrent thrombotic events still occur, and enhanced bleeding risk remains a challenge that needs to be addressed. These limitations have stimulated research interest in identifying and developing new antiplatelet targets (Figure 3).

3.1. Targeting Platelet Adhesion

3.1.1. Inhibition of Von Willebrand Factor-Glycoprotein 1bα-Mediated Platelet Activation

VWF is synthesized by endothelial cells and megakaryocytes. VWF activity depends on the size of the multimer being ultra-large VWF multimers highly reactive with platelets. The monomeric VWF displays a multi-domain structure which includes an A3 domain (interacts with exposed vascular collagen) and an A1 domain (binds to platelet GPIbα). A1 interaction with GPIbα favours platelet rolling and adhesion, especially under high share rate conditions. GPVI and integrin α2β1 further support tight platelet adhesion. The resultant platelet activation induces the conformational change of GPIIb/IIIa, which favours platelet–platelet interaction (i.e., platelet aggregation) by binding to fibrinogen (primary ligand) or to the C1 domain of VWF. Although VWF also plays a pivotal role in platelet aggregation by serving as an intercellular bridge between platelets, efforts have mainly focused on the discovery of pharmacological agents able to interfere with VWF-mediated platelet adhesion either by blocking the VWF-collagen or the VWF-GPIbα interaction. Promising preclinical and proof-of-concept clinical trials have supported their antithrombotic potential, as described below [95].
Anfibatide is a direct GPIb antagonist purified from snake (Deinagkistrodon acutus) venom that prevents GPIb interaction with VWF. Intravenous administration of anfibatide in NSTEMI patients (phase Ib/IIa study) proved feasible and safe and markedly inhibited platelet aggregation without increasing the risk of bleeding [96]. A phase II trial is currently assessing its safety and efficacy in STEMI patients before primary PCI being the primary endpoint TIMI myocardial perfusion grades (NCT02495012). In the field of stroke, administering anfibatide after cerebral ischemia/reperfusion injury in rats has been shown to significantly improve ischemic lesions alleviating inflammation and apoptosis in a dose-dependent manner [97] and preserving blood-brain barrier integrity [98]. These observations further support the contribution of platelets to inflammation and immune responses in ischemic damage beyond their function in hemostasis [99].
Caplacizumab (formerly ALX-0081) is a humanized monoclonal nanobody that targets the A1 domain of VWF, preventing its interaction with GPIb. After promising results in phase I studies (healthy subjects and stable angina patients undergoing PIC) [100], a phase II study in high-risk patients with ACS undergoing PCI (NCT01020383) is currently underway and aims to compare the safety and efficacy of caplacizumab vs. abciximab on top of standard antithrombotic therapy (ASA, clopidogrel, and heparin).
Aptamers have also been developed to block the A1 domain (Table 5). As such, BT200 has been shown to effectively block VWF activity in both ACS [101] and stroke [102] patients by binding to the VWF-A1 domain and is currently being tested in healthy volunteers (phase I, NCT04103034).

3.1.2. Glycoprotein VI: Inhibition of Collagen-Mediated Platelet Activation

GPVI is a platelet- and megakaryocyte-specific 60–65 kDa immunoglobulin-like transmembrane receptor. It is expressed at the platelet surface and is associated with the FcR γ (Fc receptor γ)-chain, which is responsible for the signaling via its immunoreceptor-tyrosine-based-activation motif. GPVI is considered the main collagen receptor in platelets, although it also binds to other substrates, including fibrin, fibrinogen, fibronectin, galectin-3, or laminin [103]. Activation of the GPVI–FcRγ complex initiates intracellular signaling through a tyrosine kinase-based signaling pathway [104] that eventually triggers calcium mobilization and the resultant platelet activation [105]. Several experimental studies have supported that GPVI seems to have little or no impact on hemostasis. As such, patients lacking functional GPVI have shown mild bleeding diathesis [106] unless they have moderate to severe thrombocytopenia [107]. Likewise, a mutation in the GPVI gene identified in the Chilean population that prevents GPVI surface expression has not been associated with a significant increase in bleeding and has been hypothesized to confer a protective benefit against CVD [108]. Overall, the fact that GPVI is uniquely expressed in platelets and megakaryocytes and has reported minor involvement in hemostasis [109,110] has made GPVI inhibition a promising approach to prevent thrombosis while limiting bleeding risk.
Revacept, a competitive antagonist to GPVI-collagen signaling, is one of the most studied drugs. Revacept is a dimeric, soluble fusion protein composed of the extracellular domain of the GPVI receptor and the human Fc-fragment. It competes with endogenous platelet GPVI for binding to exposed collagen fibers preventing platelet activation [105]. Since revacept targets the exposed vascular collagen, it does not interfere with circulating platelets beyond the atherosclerotic lesion, exerting a little effect on systemic hemostasis or bleeding as suggested in experimental models and a phase I clinical trial [111]. Revacept has been tested in phase II clinical trials [112] in patients with stable coronary heart disease undergoing PCI. Yet, no significant differences were observed in the primary endpoint (death or myocardial injury) or bleeding between the treated and placebo arm. Future studies are being planned to address its efficacy in patients at higher risk of ischemic events (e.g., in the context of ACS), where collagen-induced platelet activation may play a more important role.
In the setting of ischemic stroke, revacept is currently being tested in a phase II clinical trial (NCT01645306) in patients with symptomatic carotid artery stenosis (history of ischemic stroke, transitory ischemic attack or amaurosis fugax within the last 30 days) to check its efficacy in secondary prevention of thromboembolic ischemic events.
Several monoclonal antibodies against GPVI have also been developed, as detailed in Table 5, the most important being glenzocimab (ACT017). This monoclonal antibody binds to human GPVI and has inhibited platelet adhesion, aggregation, and thrombus formation onto collagen surface under arterial flow conditions [113]. Glenzocimab has a short plasma half-life requiring to be infused intravenously for 6 or 12 h to maintain the necessary duration of effect [114]. Glenzocimab has been demonstrated to sufficiently block collagen-induced platelet aggregation in a phase I study [115] with an excellent safety profile (no evidence of thrombocytopenia or excess bleeding). Glenzocimab is being tested in a phase II/III trial to evaluate the safety and efficacy of a single dose of glenzocimab used in combination with standard of care (thrombolysis and thrombectomy) for acute ischemic stroke (ACTIVASE NCT05070260).
Table 5. Novel antiplatelets in the preclinical phase.
Table 5. Novel antiplatelets in the preclinical phase.
AntiplateletTypeMechanism of ActionStudies Conducted So Far
TAGX-0004 (studies in vitro)AptamerVWF inhibitionIt has excellent affinity with VWF-A1 domain and a superior antithrombotic potential than ARC1779 [116].
ARC1779 (intravenous)AptamerVWF inhibitionIn a phase II trial, it reduced cerebral thromboembolism in patients undergoing carotid endarterectomy [117]. However, the study was terminated due to a lack of funding and associated increased bleeding risk. Further development of ARC1779 was halted.
AJW200 (intravenous)Monoclonal antibodyVWF inhibitionTested as adjunctive therapy with tPA in a mouse model of embolic stroke where it showed a synergistic effect and improved behavioural function [118]. In monkeys, it has been shown to inhibit high-shear-stress-induced platelet adhesion, aggregation, and thrombin generation [119].
82D6A3 (intravenous)Monoclonal antibody (A3 domain)VWF inhibitionIt has been tested in baboons, showing potent antithrombotic activities without significantly prolonging the bleeding time [120].
Caplacizumab (intravenous/subcutaneous)NanobodyVWF inhibitionApproved for the treatment of immune-mediated thrombotic thrombocytopenic purpura [121].
h6B4-Fab (intravenous)Monoclonal antibodyGPIb inhibitionReduce thrombus formation in baboons with minimal effect on bleeding time [122].
SZ2 (intravenous)Monoclonal antibodyGPIb inhibitionIn vitro, functional studies revealed that it prevents platelet adhesion to VWF under high-shear stress and inhibits ristocetin-induced platelet aggregation in a dose-dependent manner [123].
JAQ1 (Intravenous)Monoclonal antibodyGPVI inhibitionIt protects against lethal thromboembolism in mice with minimal impact on hemostasis [124,125].
SCH-28 (studies in vitro)Small moleculePAR4 inhibitionIt inhibits PAR-4-mediated platelet activation and aggregation by blocking the thrombin exosite II binding domain [126].
HPW-RX40 (intravenous)Small moleculePDI inhibitionReduces thrombus formation in whole human blood under flow conditions and protects mice from ferric chloride-induced thrombus formation [127].
ML359 (studies in vitro)Small moleculePDI InhibitionIt exerts no cytotoxicity in three human cell lines and inhibits platelet aggregation [128].
ML355 (oral)Small molecule12-Lipoxygenase inhibitionIt reduces thrombus growth and vessel occlusion in a mouse model of arterial thrombosis with minimal impact on hemostasis [129].
MIPS-9922 (intravenous)Small moleculePI3Kβ inhibitionIt prevents arterial thrombus formation in an in vivo electrolytic mouse model of thrombosis with minimal impact on hemostasis [130].
scFv (intravenous)AntibodyGPIIb/IIIa inhibitionIt has demonstrated comparable antithrombotic efficacy to currently used GPIIb/IIIa inhibitors (tirofiban and eptifibatide) in a mice model of ferric chloride-induced thrombosis with minimal impact on hemostasis [131].
mP6 (intravenous)PéptideGPIIb/IIIa inhibitionIt has proven superior to aspirin and is similar to ticagrelor in a mice model of ferric chloride-induced thrombosis with minimal effects on hemostasis [132].
SAR216471 (oral)Small moleculeP2Y12 InhibitionIt has shown potent antithrombotic activity in a rat arterio-venous shunt model with no effect on hemostasia [133].
AZD1283 (oral)Small moleculeP2Y12 InhibitionIt has shown potent antithrombotic efficacy in a rat model of ferric chloride-induced thrombosis and lowers bleeding risk compared to clopidogrel [134].
BMS-884775 (oral)Small moleculeP2Y1 InhibitionIt has demonstrated, in a rabbit model of thrombosis, similar efficacy to prasugrel with less bleeding risk [135].
MRS2500 (intravenous)Small moleculeP2Y1 InhibitionIt prevents carotid artery thrombosis in monkey models of electrolytic-mediated arterial thrombosis with a concomitant mild prolongation in bleeding time [136].
GLS-409 (intravenous)Small moleculeP2Y1 and P2Y12 InhibitionA
It attenuates thrombosis in a canine model of unstable angina and reduces platelet aggregation to a comparable extent to cangrelor or the combination of cangrelor with a selective P2Y1 inhibitor [137].
Troα6 and Troα10 (intravenous)PeptidesGPVI inhibitionIt inhibits collagen-induced platelet aggregation and thrombus formation in a ferric chloride-induced thrombosis model without prolonging bleeding time [138].
BI1002494 (oral)PeptideGPVI inhibitionIt reduces infarct sizes and improves neurological outcomes in a mouse model of cerebral ischemia without affecting hemostasis [139].
Tyrosine kinase inhibitors are also being developed to prevent downstream signaling initiated by activation of the GPVI-FcRγ complex. As such, platelet activation through GPVI relies on a potent protein tyrosine kinase cascade culminating in the activation of the tyrosine kinase Syk (spleen-associated tyrosine kinase). Tyrosine kinase inhibitors have been shown to exert antiplatelet effects in cancer patients (e.g., pazopanib in patients with renal cell carcinoma) [140] and short-term studies with ibrutinib analogs Btki (Bruton’s tyrosine kinase inhibitors) 43607 and Btki 43761 have shown a dramatic reduction in collagen-induced platelet aggregation in non-human primates without measurable effects on plasma clotting times or bleeding risk [141]. In addition, the Syk inhibitors PRT-060318 and BI1002494 have been shown to reduce thrombus stability in vitro [142] and thrombosis in a mouse model of cerebral ischemia [139], respectively. Finally, ibrutinib has also been shown to block CLEC-2-mediated platelet activation [143]. CLEC-2 is a platelet-activating type II transmembrane receptor which has a function similar to that of GPVI in activating Syk [144].

3.2. Targeting Platelet Activation

3.2.1. PAR1 and PAR4: Inhibition of Thrombin-Mediated Platelet Activation

Thrombin activates human platelets through the protease-activated receptor (PAR)-1 and PAR-4. PARs are G protein-coupled receptors whose activation by thrombin depends on proteolytic cleavage of the N-terminal domain of the receptor, generating a new amino terminus that acts as a tethered ligand to activate the receptor. PAR-4 has shown to interact with PAR-1 and P2Y12, inducing sustained platelet activation, whereas PAR-1 does not interact with ADP receptors leading to an acute platelet response. Hence, blockage of the P2Y12 receptor may suppress PAR-4-mediated platelet aggregation, while PAR-1-mediated effects remain unaltered [145].
PAR-1 has shown a high affinity for thrombin, whereas higher thrombin levels are required to activate PAR-4. Hence, PAR-1 has become the focus of intense research as a therapeutic antiplatelet target. Vorapaxar is a competitive PAR-1 antagonist that irreversibly binds to the ligand-binding pocket on the extracellular surface of PAR-1. Based on two large phases III clinical trials (TRA 2°P–TIMI 50 [146] and TRACER [147] vorapaxar may be used on top of standard antiplatelet therapy in the secondary prevention of ischemic events in patients with a history of MI or symptomatic peripheral artery disease. Yet, vorapaxar is contraindicated in patients with a history of stroke or transient ischemic attack because it has been associated with increased intracranial bleeding. However, subgroup analyses of both trials have found that vorapaxar might be potentially beneficial in patients with previous MI, diabetes, coronary artery bypass grafting, and ischemic stroke [148,149]. So far, vorapaxar has been shown to reduce thrombus formation in post-MI patients treated with potent P2Y12 inhibitors [150].
PZ-128 is a pepducin inhibitor of PAR1 for patients with CAD/ACS undergoing coronary interventions. Pepducins are lipidated peptides which target the cytoplasmic surface of their cognate receptor, not affecting the ligand-binding. PZ-128 has experimentally been demonstrated to reduce acute arterial thrombosis and atherosclerotic plaque burden [151]. Furthermore, PZ-128 was recently tested in a phase II trial in NSTEMI or stable angina patients undergoing PCI and appeared to be safe, well-tolerated, and potentially reduce periprocedural myonecrosis when administered on top of standard antiplatelet therapy [152].
As per PAR-4 inhibitors, BMS-986141 has been demonstrated to reduce platelet-rich thrombus formation under a high shear rate [153] and is currently being tested in a phase IIa trial (NCT05093790). A similar drug, BMS-986120, has been recently tested with success in humans (phase I) [154] after encouraging data from preclinical studies where it has shown robust antithrombotic activity and a low bleeding profile [155,156].

3.2.2. Inhibition of Phosphoinositide 3-Kinase Beta (PI3Kβ)

PI3Kβ is a lipid kinase that acts as an important mediator in the signal transduction downstream of the activation of P2Y12, GPIIb/IIIa, GPVI, PAR, and GPIb and plays a pivotal role in platelet aggregation and thrombus stability.
Based on the specific PI3Kβ inhibitor, TGX-221, which has only been tested in preclinical studies, a new molecule with better pharmacological properties has been developed, AZD-6482. This drug has shown in a phase I trial to moderately inhibit ADP- and collagen-induced platelet aggregation, particularly under high shear stress conditions with only mild prolonged bleeding time [157]. In another phase I study, the combination of AZD-6482 with ASA provided greater platelet inhibition compared to DAPT with ASA and clopidogrel without translating into prolonged bleeding times [158]. A new phase II trial (STARS) is planned to test the safety and tolerability of this drug in reperfusion for stroke (NCT05363397).

3.2.3. Selatogrel: The New Antagonist of the P2Y12 Receptor

Selatogrel (ACT-246475) is a new potent, reversible, and selective inhibitor of the P2Y12 platelet receptor. Its efficacy and safety have already been confirmed in phase I and II clinical trials. In contrast to the currently used P2Y12 inhibitors (i.e., oral or intravenous administration), selatogrel is administered subcutaneously, overcoming potential pharmacokinetic limitations of other P2Y12 inhibitors, including the delay of absorption and lack of enteral access for administration with oral formulations; the need for intravenous access with cangrelor; or the need for metabolization (e.g., clopidogrel and prasugrel) to be ideal in the critical 3-h window during an ACS [159]. Additionally, selatogrel seems to have a lower bleeding risk profile than clopidogrel or ticagrelor. A study performed in mice showed that the stability of hemostatic seals was undisturbed in the presence of selatogrel, unlike clopidogrel or ticagrelor. The authors suggested that the mechanism underlying the differences in blood loss profiles among these P2Y12 receptor antagonists was related to off-target interference with endothelial and neutrophil cells and fibrin-mediated stabilization of hemostatic seals [160]. Subsequently, phase I and phase II clinical trials have confirmed that selatogrel provides sustained and reversible P2Y12 platelet inhibition with an acceptable safety profile [159]. A phase III clinical trial is currently underway (NCT04957719).

3.2.4. New P2Y1 Receptor Antagonists

Besides the P2Y12 receptor, human platelets express another purinergic ADP receptor named P2Y1. The binding of ADP to P2Y1 initiates platelet aggregation response which may be reverted, while P2Y12 activation leads to irreversible platelet aggregation. Therefore, complete platelet aggregation requires a complex interplay and coactivation of both P2Y1 and P2Y12 receptors [161]. Following this assumption, several P2Y1 inhibitors have been developed, though so far, they have only been tested in animal models, as detailed in Table 5 [162].

3.3. Targeting Platelet Aggregation and Thrombus Propagation

3.3.1. New Inhibitor of GP IIb/IIIa

Zalunfiban (RUC-4) is a second-generation small-molecule platelet GPIIb/IIIa inhibitor that blocks the receptor in its inactive conformation. This blockade avoids the drug-induced thrombocytopenia associated with other GPIIb/IIIa inhibitors since it prevents the exposition of epitopes that are potential targets for thrombocytopenia-related antibodies. Subcutaneous administration of RUC-4 in healthy subjects and stable coronary artery disease patients on ASA (Phase I trial) has shown a rapid (<15 min), potent (>80% reduction of platelet aggregation), and reversible (platelet function is restored after 1–2 h) platelet inhibitory effect [163]. These observations were confirmed in a phase IIa trial in the setting of STEMI [164]. Currently, RUC-4 is being tested in phase IIb trial in STEMI patients undergoing primary PCI (NCT04825743). Other GPIIb/IIIa inhibitors have been developed and are currently being tested in the preclinical setting, as detailed in Table 5.

3.3.2. Inhibition of Protein Disulfide Isomerase (PDI)

PDI is an enzyme in the endoplasmic reticulum that catalyzes the modification of thiol-disulfide bonds during protein synthesis and is also expressed on the surface of multiple cells, including platelets. Four members of the PDI family of enzymes, including PDI, ERp57, ERp72, and ERp5, are secreted from activated platelets and endothelial cells at the site of vascular injury. The mechanisms by which extracellular PDI regulates platelet function remain to be determined. However, it is thought to interact with prothrombotic components, including GPIIb/IIIa, α2β1, vWF, GPIbα, and TF supporting, and thus, platelet activation, aggregation, and coagulation [165,166,167].
Quercetin flavonoids (mainly isoquercetin) are potent PDI inhibitors present in fruits and vegetables. They have been tested primarily in the field of cancer and venous thromboembolism where it has been shown, in phase II trial, to improve hypercoagulability in advanced cancer [168]. However, its potential role in the context of CVD has yet to be established [169,170]. So far, other PDI inhibitors are in the pipeline since they have been shown to exert antithrombotic effects in vitro and in experimental animal models (Table 5).

4. Conclusions

Despite the major advances in antithrombotic therapy accomplished over the last decades, atherothrombotic events remain a leading cause of death worldwide. The secondary prevention of both ischemic heart disease and ischemic stroke requires effective antiplatelets and anticoagulants without bleeding side effects. Research conducted over the last years has led to a deeper understanding of the molecular mechanisms regulating atherothrombosis and hemostasis, providing new targets for intervention [5,6,171,172,173]. New antithrombotic strategies have been developed and assessed in preclinical animal models, and some have already reached clinical testing. As per the coagulation cascade, new anticoagulants have focused on the intrinsic coagulation pathway to prevent ischemic coronary and cerebral events. In this regard, although the long journey from animal studies to randomized clinical trials has just started, hopefully, some of these promising strategies will reach routine clinical use, providing the patient with optimal protection against arterial thrombosis inhibition while preserving hemostasis.

Author Contributions

I.B. and G.V. wrote the paper. F.W. provided an in-depth and constructive review of the content. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Grant PID2021-128891OB-I00 (to G.V.) and PLEC2021- 007664- NextGenerationEU (to G.V.) funded by MCIN/AEI/10.13039/501100011033 and Fondo Europeo de Desarrollo Regional (FEDER) A way of making Europe; and the SEC/FEC-INV-TRL 20/015 funded by the Spanish Society of Cardiology (to G.V.) We thank the Fundación Investigación Cardiovascular—Fundación Jesus Serra for their continuous support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work is part of the Autonomous University of Barcelona requirement for the Doctorate in Medicine (Ignacio Barriuso).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cardiovascular Diseases (CVDs) [Internet]. Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 24 September 2022).
  2. Roth, G.A.; Forouzanfar, M.H.; Moran, A.E.; Barber, R.; Nguyen, G.; Feigin, V.L.; Naghavi, M.; Mensah, G.A.; Murray, C.J. Demographic and Epidemiologic Drivers of Global Cardiovascular Mortality. N. Engl. J. Med. 2015, 372, 1333–1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Global Burden of Disease Collaborative Network. Global Burden of Disease Study 2019 (GBD 2019) Results; Institute for Health Metrics and Evaluation (IHME): Seattle, WA, USA, 2019; Available online: http://ghdx.healthdata.org/gbd-results-tool (accessed on 15 September 2022).
  4. Vilahur, G.; Badimon, J.J.; Bugiardini, R.; Badimon, L. Perspectives: The burden of cardiovascular risk factors and coronary heart disease in Europe and worldwide. Eur. Heart J. Suppl. 2014, 16, A7–A11. [Google Scholar] [CrossRef] [Green Version]
  5. Badimon, L.; Storey, R.F.; Vilahur, G. Update on lipids, inflammation and atherothrombosis. Thromb. Haemost. 2011, 105, 34–42. [Google Scholar]
  6. Patrono, C.; Morais, J.; Baigent, C.; Collet, J.P.; Fitzgerald, D.; Halvorsen, S.; Rocca, B.; Siegbahn, A.; Storey, R.F.; Vilahur, G. Antiplatelet Agents for the Treatment and Prevention of Coronary Atherothrombosis. J. Am. Coll. Cardiol. 2017, 70, 1760–1776. [Google Scholar] [CrossRef] [PubMed]
  7. Badimon, L.; Vilahur, G. Platelets, arterial thrombosis and cerebral ischemia. Cerebrovasc. Dis. 2007, 24, 30–39. [Google Scholar] [CrossRef] [PubMed]
  8. Badimon, L.; Padró, T.; Vilahur, G. Atherosclerosis, platelets and thrombosis in acute ischaemic heart disease. Eur. Heart J. Acute Cardiovasc. Care 2012, 1, 60–74. [Google Scholar] [CrossRef] [Green Version]
  9. Parker, W.A.E.; Gorog, D.A.; Geisler, T.; Vilahur, G.; Sibbing, D.; Rocca, B.; Storey, R.F. Prevention of stroke in patients with chronic coronary syndromes or peripheral arterial disease. Eur. Heart J. Suppl. 2020, 22, M26–M34. [Google Scholar] [CrossRef]
  10. Hindricks, G.; Potpara, T.; Dagres, N.; Arbelo, E.; Bax, J.J.; Blomström-Lundqvist, C.; Boriani, G.; Castella, M.; Dan, G.-A.; Dilaveris, P.E.; et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur. Heart J. 2021, 42, 373–498. [Google Scholar]
  11. Díaz-Guzmán, J.; Freixa-Pamias, R.; García-Alegría, J.; Cabeza, A.-I.P.; Roldán-Rabadán, I.; Antolin-Fontes, B.; Rebollo, P.; Llorac, A.; Genís-Gironés, M.; Escobar-Cervantes, C. Epidemiology of atrial fibrillation-related ischemic stroke and its association with DOAC uptake in Spain: First national population-based study 2005 to 2018. Rev. Esp. Cardiol. 2022, 75, 496–505. [Google Scholar] [CrossRef]
  12. Cha, M.-J.; Choi, E.-K.; Han, K.-D.; Lee, S.-R.; Lim, W.-H.; Oh, S.; Lip, G.Y.H. Effectiveness and Safety of Non-Vitamin K Antagonist Oral Anticoagulants in Asian Patients with Atrial Fibrillation. Stroke 2017, 48, 3040–3048. [Google Scholar] [CrossRef]
  13. Dhakal, P.; Rayamajhi, S.; Verma, V.; Gundabolu, K.; Bhatt, V.R. Reversal of Anticoagulation and Management of Bleeding in Patients on Anticoagulants. Clin. Appl. Thromb. Hemost. 2017, 23, 410–415. [Google Scholar] [CrossRef]
  14. Desai, N.R.; Cornutt, D. Reversal agents for direct oral anticoagulants: Considerations for hospital physicians and intensivists. Hosp. Pract. 2019, 47, 113–122. [Google Scholar] [CrossRef]
  15. Jourdi, G.; Le Bonniec, B.; Gouin-Thibault, I. Strategies of neutralization of the direct oral anticoagulants effect: Review of the literature. Ann. Biol. Clin. 2019, 77, 67–78. [Google Scholar] [CrossRef]
  16. Ansell, J.; Laulicht, B.E.; Bakhru, S.H.; Burnett, A.; Jiang, X.; Chen, L.; Baker, C.; Villano, S.; Steiner, S. Ciraparantag, an anticoagulant reversal drug: Mechanism of action, pharmacokinetics, and reversal of anticoagulants. Blood 2021, 137, 115–125. [Google Scholar] [CrossRef]
  17. Ansell, J.E.; Bakhru, S.H.; Laulicht, B.E.; Steiner, S.S.; Grosso, M.; Brown, K.; Dishy, V.; Noveck, R.J.; Costin, J.C. Use of PER977 to reverse the anticoagulant effect of edoxaban. N. Engl. J. Med. 2014, 371, 2141–2142. [Google Scholar] [CrossRef]
  18. Sheffield, W.P.; Lambourne, M.D.; Eltringham-Smith, L.J.; Bhakta, V.; Arnold, D.M.; Crowther, M.A. γT-S195A thrombin reduces the anticoagulant effects of dabigatran in vitro and in vivo. J. Thromb. Haemost. 2014, 12, 1110–1115. [Google Scholar] [CrossRef]
  19. Jourdi, G.; Abdoul, J.; Siguret, V.; Decleves, X.; Frezza, E.; Pailleret, C.; Gouin-Thibault, I.; Gandrille, S.; Neveux, N.; Samama, C.M.; et al. Induced forms of α2-macroglobulin neutralize heparin and direct oral anticoagulant effects. Int. J. Biol. Macromol. 2021, 184, 209–217. [Google Scholar] [CrossRef]
  20. Chan, N.; Sobieraj-Teague, M.; Eikelboom, J.W. Direct oral anticoagulants: Evidence and unresolved issues. Lancet 2020, 396, 1767–1776. [Google Scholar] [CrossRef]
  21. Eikelboom, J.W.; Connolly, S.J.; Brueckmann, M.; Granger, C.B.; Kappetein, A.P.; Mack, M.J.; Blatchford, J.; Devenny, K.; Friedman, J.; Guiver, K.; et al. Dabigatran versus warfarin in patients with mechanical heart valves. N. Engl. J. Med. 2013, 369, 1206–1214. [Google Scholar] [CrossRef] [Green Version]
  22. Pengo, V.; Denas, G.; Zoppellaro, G.; Jose, S.P.; Hoxha, A.; Ruffatti, A.; Andreoli, L.; Tincani, A.; Cenci, C.; Prisco, D.; et al. Rivaroxaban vs warfarin in high-risk patients with antiphospholipid syndrome. Blood 2018, 132, 1365–1371. [Google Scholar] [CrossRef]
  23. Merlini, P.A.; Bauer, K.A.; Oltrona, L.; Ardissino, D.; Cattaneo, M.; Belli, C.; Mannucci, P.M.; Rosenberg, R.D. Persistent activation of coagulation mechanism in unstable angina and myocardial infarction. Circulation 1994, 90, 61–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Rothberg, M.B.; Celestin, C.; Fiore, L.D.; Lawler, E.; Cook, J.R. Warfarin plus aspirin after myocardial infarction or the acute coronary syndrome: Meta-analysis with estimates of risk and benefit. Ann. Intern. Med. 2005, 143, 241–250. [Google Scholar] [CrossRef] [PubMed]
  25. Eikelboom, J.W.; Connolly, S.J.; Bosch, J.; Dagenais, G.R.; Hart, R.G.; Shestakovska, O.; Diaz, R.; Alings, M.; Lonn, E.M.; Anand, S.S.; et al. Rivaroxaban with or without Aspirin in Stable Cardiovascular Disease. N. Engl. J. Med. 2017, 377, 1319–1330. [Google Scholar] [CrossRef] [PubMed]
  26. Mega, J.L.; Braunwald, E.; Wiviott, S.D.; Bassand, J.P.; Bhatt, D.L.; Bode, C.; Burton, P.; Cohen, M.; Cook-Bruns, N.; Fox, K.A.; et al. Rivaroxaban in patients with a recent acute coronary syndrome. N. Engl. J. Med. 2012, 366, 9–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Wallentin, L.; Wilcox, R.G.; Weaver, W.D.; Emanuelsson, H.; Goodvin, A.; Nyström, P.; Bylock, A.; ESTEEM Investigators. Oral ximelagatran for secondary prophylaxis after myocardial infarction: The ESTEEM randomised controlled trial. Lancet 2003, 362, 789–797. [Google Scholar] [CrossRef]
  28. Wheeler, A.P.; Gailani, D. The Intrinsic Pathway of Coagulation as a Target for Antithrombotic Therapy. Hematol. Oncol. Clin. N. Am. 2016, 30, 1099–1114. [Google Scholar] [CrossRef] [Green Version]
  29. Schmaier, A.H.; Stavrou, E.X. Factor XII—What’s important but not commonly thought about. Res. Pract. Thromb. Haemost. 2019, 3, 599–606. [Google Scholar] [CrossRef] [Green Version]
  30. Heestermans, M.; Naudin, C.; Mailer, R.K.; Konrath, S.; Klaetschke, K.; Jämsä, A.; Frye, M.; Deppermann, C.; Pula, G.; Kuta, P.; et al. Identification of the factor XII contact activation site enables sensitive coagulation diagnostics. Nat. Commun. 2021, 12, 5596. [Google Scholar] [CrossRef]
  31. Srivastava, P.; Gailani, D. The rebirth of the contact pathway: A new therapeutic target. Curr. Opin. Hematol. 2020, 27, 311–319. [Google Scholar] [CrossRef]
  32. Kalinin, D.V. Factor XII(a) inhibitors: A review of the patent literature. Expert Opin. Ther. Pat. 2021, 31, 1155–1176. [Google Scholar] [CrossRef]
  33. Fredenburgh, J.C.; Weitz, J.I. New anticoagulants: Moving beyond the direct oral anticoagulants. J. Thromb. Haemost. 2021, 19, 20–29. [Google Scholar] [CrossRef]
  34. Craig, T.; Magerl, M.; Levy, D.S.; Reshef, A.; Lumry, W.R.; Martinez-Saguer, I.; Jacobs, J.S.; Yang, W.H.; Ritchie, B.; Aygören-Pürsün, E.; et al. Prophylactic use of an anti-activated factor XII monoclonal antibody, garadacimab, for patients with C1-esterase inhibitor-deficient hereditary angioedema: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2022, 399, 945–955. [Google Scholar] [CrossRef]
  35. McKenzie, A.; Roberts, A.; Malandkar, S.; Feuersenger, H.; Panousis, C.; Pawaskar, D. A phase I, first-in-human, randomized dose-escalation study of anti-activated factor XII monoclonal antibody garadacimab. Clin. Transl. Sci. 2022, 15, 626–637. [Google Scholar] [CrossRef]
  36. Larsson, M.; Rayzman, V.; Nolte, M.W.; Nickel, K.F.; Björkqvist, J.; Jämsä, A.; Hardy, M.P.; Fries, M.; Schmidbauer, S.; Hedenqvist, P.; et al. A factor XIIa inhibitory antibody provides thromboprotection in extracorporeal circulation without increasing bleeding risk. Sci. Transl. Med. 2014, 6, 222ra17. [Google Scholar] [CrossRef] [Green Version]
  37. Worm, M.; Köhler, E.C.; Panda, R.; Long, A.; Butler, L.M.; Stavrou, E.X.; Nickel, K.F.; Fuchs, T.A.; Renné, T. The factor XIIa blocking antibody 3F7: A safe anticoagulant with anti-inflammatory activities. Ann. Transl. Med. 2015, 3, 247. [Google Scholar] [CrossRef]
  38. Matafonov, A.; Leung, P.Y.; Gailani, A.E.; Grach, S.L.; Puy, C.; Cheng, Q.; Sun, M.F.; McCarty, O.J.; Tucker, E.I.; Kataoka, H.; et al. Factor XII inhibition reduces thrombus formation in a primate thrombosis model. Blood 2014, 123, 1739–1746. [Google Scholar] [CrossRef] [Green Version]
  39. Wallisch, M.; Lorentz, C.U.; Lakshmanan, H.H.S.; Johnson, J.; Carris, M.R.; Puy, C.; Gailani, D.; Hinds, M.T.; McCarty, O.J.T.; Gruber, A.; et al. Antibody inhibition of contact factor XII reduces platelet deposition in a model of extracorporeal membrane oxygenator perfusion in nonhuman primates. Res. Pract. Thromb. Haemost. 2020, 4, 205–216. [Google Scholar] [CrossRef] [Green Version]
  40. Pireaux, V.; Tassignon, J.; Demoulin, S.; Derochette, S.; Borenstein, N.; Ente, A.; Fiette, L.; Douxfils, J.; Lancellotti, P.; Guyaux, M.; et al. Anticoagulation with an Inhibitor of Factors XIa and XIIa During Cardiopulmonary Bypass. J. Am. Coll. Cardiol. 2019, 74, 2178–2189. [Google Scholar] [CrossRef]
  41. Demoulin, S.; Godfroid, E.; Hermans, C. Dual inhibition of factor XIIa and factor XIa as a therapeutic approach for safe thromboprotection. J. Thromb. Haemost. 2021, 19, 323–329. [Google Scholar] [CrossRef]
  42. Decrem, Y.; Rath, G.; Blasioli, V.; Cauchie, P.; Robert, S.; Beaufays, J.; Frère, J.M.; Feron, O.; Dogné, J.M.; Dessy, C.; et al. Ir-CPI, a coagulation contact phase inhibitor from the tick Ixodes ricinus, inhibits thrombus formation without impairing hemostasis. J. Exp. Med. 2009, 206, 2381–2395. [Google Scholar] [CrossRef] [Green Version]
  43. Yau, J.W.; Liao, P.; Fredenburgh, J.C.; Stafford, A.R.; Revenko, A.S.; Monia, B.P.; Weitz, J.I. Selective depletion of factor XI or factor XII with antisense oligonucleotides attenuates catheter thrombosis in rabbits. Blood 2014, 123, 2102–2107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Liu, J.; Cooley, B.C.; Akinc, A.; Butler, J.; Borodovsky, A. Knockdown of liver-derived factor XII by GalNAc-siRNA ALN-F12 prevents thrombosis in mice without impacting hemostatic function. Thromb. Res. 2020, 196, 200–205. [Google Scholar] [CrossRef] [PubMed]
  45. May, F.; Krupka, J.; Fries, M.; Thielmann, I.; Pragst, I.; Weimer, T.; Panousis, C.; Nieswandt, B.; Stoll, G.; Dickneite, G.; et al. FXIIa inhibitor rHA-Infestin-4: Safe thromboprotection in experimental venous, arterial and foreign surface-induced thrombosis. Br. J. Haematol. 2016, 173, 769–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Krupka, J.; May, F.; Weimer, T.; Pragst, I.; Kleinschnitz, C.; Stoll, G.; Panousis, C.; Dickneite, G.; Nolte, M.W. The Coagulation Factor XIIa Inhibitor rHA-Infestin-4 Improves Outcome after Cerebral Ischemia/Reperfusion Injury in Rats. PLoS ONE 2016, 11, e0146783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Hopp, S.; Albert-Weissenberger, C.; Mencl, S.; Bieber, M.; Schuhmann, M.K.; Stetter, C.; Nieswandt, B.; Schmidt, P.M.; Monoranu, C.M.; Alafuzoff, I.; et al. Targeting coagulation factor XII as a novel therapeutic option in brain trauma. Ann. Neurol. 2016, 79, 970–982. [Google Scholar] [CrossRef]
  48. Salomon, O.; Steinberg, D.M.; Zucker, M.; Varon, D.; Zivelin, A.; Seligsohn, U. Patients with severe factor XI deficiency have a reduced incidence of deep-vein thrombosis. Thromb. Haemost. 2011, 105, 269–273. [Google Scholar] [CrossRef]
  49. Tucker, E.I.; Marzec, U.M.; White, T.C.; Hurst, S.; Rugonyi, S.; McCarty, O.J.; Gailani, D.; Gruber, A.; Hanson, S.R. Prevention of vascular graft occlusion and thrombus-associated thrombin generation by inhibition of factor XI. Blood 2009, 113, 936–944. [Google Scholar] [CrossRef] [Green Version]
  50. Salomon, O.; Steinberg, D.M.; Koren-Morag, N.; Tanne, D.; Seligsohn, U. Reduced incidence of ischemic stroke in patients with severe factor XI deficiency. Blood 2008, 111, 4113–4117. [Google Scholar] [CrossRef] [Green Version]
  51. Preis, M.; Hirsch, J.; Kotler, A.; Zoabi, A.; Stein, N.; Rennert, G.; Saliba, W. Factor XI deficiency is associated with lower risk for cardiovascular and venous thromboembolism events. Blood 2017, 129, 1210–1215. [Google Scholar] [CrossRef] [Green Version]
  52. James, P.; Salomon, O.; Mikovic, D.; Peyvandi, F. Rare bleeding disorders—Bleeding assessment tools, laboratory aspects and phenotype and therapy of FXI deficiency. Haemophilia 2014, 20, 71–75. [Google Scholar] [CrossRef]
  53. Nourse, J.; Danckwardt, S. A novel rationale for targeting FXI: Insights from the hemostatic microRNA targetome for emerging anticoagulant strategies. Pharmacol. Ther. 2021, 218, 107676. [Google Scholar] [CrossRef]
  54. Weitz, J.I.; Bauersachs, R.; Becker, B.; Berkowitz, S.D.; Freitas, M.C.S.; Lassen, M.R.; Metzig, C.; Raskob, G.E. Effect of Osocimab in Preventing Venous Thromboembolism Among Patients Undergoing Knee Arthroplasty: The FOXTROT Randomized Clinical Trial. JAMA 2020, 323, 130–139. [Google Scholar] [CrossRef]
  55. Verhamme, P.; Yi, B.A.; Segers, A.; Salter, J.; Bloomfield, D.; Büller, H.R.; Raskob, G.E.; Weitz, J.I. Abelacimab for Prevention of Venous Thromboembolism. N. Engl. J. Med. 2021, 385, 609–617. [Google Scholar] [CrossRef]
  56. Lorentz, C.U.; Tucker, E.I.; Verbout, N.G.; Shatzel, J.J.; Olson, S.R.; Markway, B.D.; Wallisch, M.; Ralle, M.; Hinds, M.T.; McCarty, O.J.T.; et al. The contact activation inhibitor AB023 in heparin-free hemodialysis: Results of a randomized phase 2 clinical trial. Blood 2021, 138, 2173–2184. [Google Scholar] [CrossRef]
  57. Cheng, Q.; Tucker, E.I.; Pine, M.S.; Sisler, I.; Matafonov, A.; Sun, M.F.; White-Adams, T.C.; Smith, S.A.; Hanson, S.R.; McCarty, O.J.; et al. A role for factor XIIa-mediated factor XI activation in thrombus formation in vivo. Blood 2010, 116, 3981–3989. [Google Scholar] [CrossRef] [Green Version]
  58. Van Montfoort, M.L.; Knaup, V.L.; Marquart, J.A.; Bakhtiari, K.; Castellino, F.J.; Hack, C.E.; Meijers, J.C. Two novel inhibitory anti-human factor XI antibodies prevent cessation of blood flow in a murine venous thrombosis model. Thromb. Haemost. 2013, 110, 1065–1073. [Google Scholar] [CrossRef] [Green Version]
  59. Hayward, N.J.; Goldberg, D.I.; Morrel, E.M.; Friden, P.M.; Bokesch, P.M. Abstract 13747: Phase 1a/1b Study of EP-7041: A novel, potent, selective, small molecule FXIa inhibitor. Circulation 2017, 136, A13747. [Google Scholar]
  60. Weitz, J.I.; Strony, J.; Ageno, W.; Gailani, D.; Hylek, E.M.; Lassen, M.R.; Mahaffey, K.W.; Notani, R.S.; Roberts, R.; Segers, A.; et al. Milvexian for the Prevention of Venous Thromboembolism. N. Engl. J. Med. 2021, 385, 2161–2172. [Google Scholar] [CrossRef]
  61. Wong, P.C.; Crain, E.J.; Bozarth, J.M.; Wu, Y.; Dilger, A.K.; Wexler, R.R.; Ewing, W.R.; Gordon, D.; Luettgen, J.M. Milvexian, an orally bioavailable, small-molecule, reversible, direct inhibitor of factor XIa: In vitro studies and in vivo evaluation in experimental thrombosis in rabbits. J. Thromb. Haemost. 2022, 20, 399–408. [Google Scholar] [CrossRef]
  62. Bristol Myers Squibb—Late-Breaking Results from Phase 2 AXIOMATIC-SSP Study of Milvexian, an Investigational OralFactor XIa Inhibitor, Show Favorable Antithrombotic Profile in Combination with Dual Antiplatelet Therap. Available online: https://news.bms.com/news/details/2022/Late-Breaking-Results-From-Phase-2-AXIOMATIC-SSP-Study-of-Milvexian-an-Investigational-Oral-Factor-XIa-Inhibitor-Show-Favorable-Antithrombotic-Profile-in-Combination-With-Dual-Antiplatelet-Therapy/default.aspx (accessed on 29 September 2022).
  63. Piccini, J.P.; Caso, V.; Connolly, S.J.; Fox, K.A.A.; Oldgren, J.; Jones, W.S.; Gorog, D.A.; Durdil, V.; Viethen, T.; Neumann, C.; et al. Safety of the oral factor XIa inhibitor asundexian compared with apixaban in patients with atrial fibrillation (PACIFIC-AF): A multicentre, randomised, double-blind, double-dummy, dose-finding phase 2 study. Lancet 2022, 399, 1383–1390. [Google Scholar] [CrossRef]
  64. Rao, S.V.; Kirsch, B.; Bhatt, D.L.; Budaj, A.; Coppolecchia, R.; Eikelboom, J.; James, S.K.; Jones, W.S.; Merkely, B.; Keller, L.; et al. A Multicenter, Phase 2, Randomized, Placebo-Controlled, Double-Blind, Parallel-Group, Dose-Finding Trial of the Oral Factor XIa Inhibitor Asundexian to Prevent Adverse Cardiovascular Outcomes Following Acute Myocardial Infarction. Circulation 2022, 146, 1196–1206. [Google Scholar] [CrossRef] [PubMed]
  65. Shoamanesh, A.; Mundl, H.; Smith, E.E.; Masjuan, J.; Milanov, I.; Hirano, T.; Agafina, A.; Campbell, B.; Caso, V.; Mas, J.-L.; et al. Factor XIa inhibition with asundexian after acute non-cardioembolic ischaemic stroke (PACIFIC-Stroke): An international, randomised, double-blind, placebo-controlled, phase 2b trial. Lancet 2022, 400, 997–1007. [Google Scholar] [CrossRef]
  66. Perera, V.; Luettgen, J.M.; Wang, Z.; Frost, C.E.; Yones, C.; Russo, C.; Lee, J.; Zhao, Y.; LaCreta, F.P.; Ma, X.; et al. First-in-human study to assess the safety, pharmacokinetics and pharmacodynamics of BMS-962212, a direct, reversible, smallmolecule factor XIa inhibitor in non-Japaneseand Japanese healthy subjects. Br. J. Clin. Pharmacol. 2018, 84, 876–887. [Google Scholar] [CrossRef] [PubMed]
  67. Beale, D.; Dennison, J.; Boyce, M.; Mazzo, F.; Honda, N.; Smith, P.; Bruce, M. ONO-7684 a novel oral FXIa inhibitor: Safety, tolerability, pharmacokinetics and pharmacodynamics in a first-in-human study. Br. J. Clin. Pharmacol. 2021, 87, 3177–3189. [Google Scholar] [CrossRef] [PubMed]
  68. Wong, P.C.; Quan, M.L.; Watson, C.A.; Crain, E.J.; Harpel, M.R.; Rendina, A.R.; Luettgen, J.M.; Wexler, R.R.; Schumacher, W.A.; Seiffert, D.A. In vitro, antithrombotic and bleeding time studies of BMS-654457, a small-molecule, reversible and direct inhibitor of factor XIa. J. Thromb. Thrombolysis 2015, 40, 416–423. [Google Scholar] [CrossRef]
  69. Kouyama, S.; Ono, T.; Hagio, T.; Sakimoto, S.; Miyata, H.; Tanaka, M.; Koda, T.; Tanaka, K.; Yanagida, D.; Sakai, M.; et al. Discovery of ONO-5450598, a highly orally bioavailable small molecule factor XIa inhibitor: The pharmacokinetic and pharmacological profiles. Res. Pract. Thromb. Haemost. 2017, 1, 99. [Google Scholar]
  70. Wong, P.C.; Crain, E.J.; Watson, C.A.; Schumacher, W.A. A small-molecule factor XIa inhibitor produces antithrombotic efficacy with minimal bleeding time prolongation in rabbits. J. Thromb. Thrombolysis 2011, 32, 129–137. [Google Scholar] [CrossRef]
  71. Büller, H.R.; Bethune, C.; Bhanot, S.; Gailani, D.; Monia, B.P.; Raskob, G.E.; Segers, A.; Verhamme, P.; Weitz, J.I. Factor XI antisense oligonucleotide for prevention of venous thrombosis. N. Engl. J. Med. 2015, 372, 232–240. [Google Scholar] [CrossRef] [Green Version]
  72. Smiley, D.A.; Becker, R.C. Factor IXa as a target for anticoagulation in thrombotic disorders and conditions. Drug Discov. Today 2014, 19, 1445–1453. [Google Scholar] [CrossRef]
  73. Vavalle, J.P.; Cohen, M.G. The REG1 anticoagulation system: A novel actively controlled factor IX inhibitor using RNA aptamer technology for treatment of acute coronary syndrome. Future Cardiol. 2012, 8, 371–382. [Google Scholar] [CrossRef] [Green Version]
  74. Afosah, D.K.; Ofori, E.; Mottamal, M.; Al-Horani, R.A. Factor IX(a) inhibitors: An updated patent review (2003–present). Expert Opin. Ther. Pat. 2022, 32, 381–400. [Google Scholar] [CrossRef]
  75. Povsic, T.J.; Vavalle, J.P.; Aberle, L.H.; Kasprzak, J.D.; Cohen, M.G.; Mehran, R.; Bode, C.; Buller, C.E.; Montalescot, G.; Cornel, J.H.; et al. A Phase 2, randomized, partially blinded, active-controlled study assessing the efficacy and safety of variable anticoagulation reversal using the REG1 system in patients with acute coronary syndromes: Results of the RADAR trial. Eur. Heart J. 2013, 34, 2481–2489. [Google Scholar] [CrossRef] [Green Version]
  76. Lincoff, A.M.; Mehran, R.; Povsic, T.J.; Zelenkofske, S.L.; Huang, Z.; Armstrong, P.W.; Steg, P.G.; Bode, C.; Cohen, M.G.; Buller, C.; et al. Effect of the REG1 anticoagulation system versus bivalirudin on outcomes after percutaneous coronary intervention (REGULATE-PCI): A randomised clinical trial. Lancet 2016, 387, 349–356. [Google Scholar] [CrossRef]
  77. Staudacher, D.L.; Putz, V.; Heger, L.; Reinöhl, J.; Hortmann, M.; Zelenkofske, S.L.; Becker, R.C.; Rusconi, C.P.; Bode, C.; Ahrens, I. Direct factor IXa inhibition with the RNA-aptamer pegnivacogin reduces platelet reactivity in vitro and residual platelet aggregation in patients with acute coronary syndromes. Eur. Heart J. Acute Cardiovasc. Care 2019, 8, 520–526. [Google Scholar] [CrossRef]
  78. Chow, F.; Benincosa, L.J.; Sheth, S.B.; Wilson, D.; Davis, C.; Minthorn, E.A.; Jusko, W.J. Pharmacokinetic and pharmacodynamic modeling of humanized anti-factor IX antibody (SB 249417) in humans. Clin. Pharmacol. Ther. 2002, 71, 235–245. [Google Scholar] [CrossRef]
  79. Eriksson, B.I.; Dahl, O.E.; Lassen, M.R.; Ward, D.P.; Rothlein, R.; Davis, G.; Turpie, A.G.G.; Fixit Study Group. Partial factor IXa inhibition with TTP889 for prevention of venous thromboembolism: An exploratory study. J. Thromb. Haemost. 2008, 6, 457–463. [Google Scholar] [CrossRef]
  80. Badimon, L.; Vilahur, G.; Rocca, B.; Patrono, C. The Key Contribution of Platelet and Vascular Arachidonic Acid Metabolism to the Pathophysiology of Atherothrombosis. Cardiovasc. Res. 2021, 117, 2001–2015. Available online: https://pubmed.ncbi.nlm.nih.gov/33484117/ (accessed on 15 September 2022). [CrossRef]
  81. Ibanez, B.; James, S.; Agewall, S.; Antunes, M.J.; Bucciarelli-Ducci, C.; Bueno, H.; Caforio, A.L.P.; Crea, F.; Goudevenos, J.A.; Halvorsen, S.; et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation. Eur. Heart J. 2018, 39, 119–177. [Google Scholar] [CrossRef] [Green Version]
  82. Knuuti, J.; Wijns, W.; Saraste, A.; Capodanno, D.; Barbato, E.; Funck-Brentano, C.; Prescott, E.; Storey, R.F.; Deaton, C.; Cuisset, T.; et al. 2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes. Eur. Heart J. 2020, 41, 407–477. [Google Scholar] [CrossRef] [Green Version]
  83. Collet, J.P.; Thiele, H.; Barbato, E.; Barthélémy, O.; Bauersachs, J.; Bhatt, D.L.; Dendale, P.; Dorobantu, M.; Edvardsen, T.; Folliguet, T.; et al. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevationThe Task Force for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation of the European Society of Cardiology (ESC). Eur. Heart J. 2021, 42, 1289–1367. [Google Scholar]
  84. Walker, J.; Cattaneo, M.; Badimon, L.; Agnelli, G.; Chan, A.T.; Lanas, A.; Rocca, B.; Rothwell, P.; Patrignani, P.; Langley, R.; et al. Highlights from the 2019 International Aspirin Foundation Scientific Conference, Rome, 28 June 2019: Benefits and risks of antithrombotic therapy for cardiovascular disease prevention. Ecancermedicalscience 2020, 14, 998. [Google Scholar] [CrossRef] [PubMed]
  85. Mehta, S.R.; Yusuf, S.; Peters, R.J.; Bertrand, M.E.; Lewis, B.S.; Natarajan, M.K.; Malmberg, K.; Rupprecht, H.; Zhao, F.; Chrolavicius, S.; et al. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: The PCI-CURE study. Lancet 2001, 358, 527–533. [Google Scholar] [CrossRef]
  86. Wiviott, S.D.; Braunwald, E.; McCabe, C.H.; Montalescot, G.; Ruzyllo, W.; Gottlieb, S.; Neumann, F.J.; Ardissino, D.; De Servi, S.; Murphy, S.A.; et al. Prasugrel versus Clopidogrel in Patients with Acute Coronary Syndromes. N. Engl. J. Med. 2007, 357, 2001–2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wallentin, L.; Becker, R.C.; Budaj, A.; Cannon, C.P.; Emanuelsson, H.; Held, C.; Horrow, J.; Husted, S.; James, S.; Katus, H.; et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N. Engl. J. Med. 2009, 361, 1045–1057. [Google Scholar] [CrossRef] [PubMed]
  88. Schrör, K.; Siller-Matula, J.M.; Huber, K. Pharmacokinetic basis of the antiplatelet action of prasugrel. Fundam. Clin. Pharmacol. 2012, 26, 39–46. [Google Scholar] [CrossRef]
  89. Li, J.; Vootukuri, S.; Shang, Y.; Negri, A.; Jiang, J.-K.; Nedelman, M.; Diacovo, T.G.; Filizola, M.; Thomas, C.J.; Coller, B.S. A novel αiIbβ3 antagonist for prehospital therapy of myocardial infarction. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 2321–2329. [Google Scholar] [CrossRef] [Green Version]
  90. Xu, Q.; Yin, J.; Si, L.Y. Efficacy and safety of early versus late glycoprotein IIb/IIIa inhibitors for PCI. Int. J. Cardiol. 2013, 162, 210–219. [Google Scholar] [CrossRef]
  91. Kleindorfer, D.O.; Towfighi, A.; Chaturvedi, S.; Cockroft, K.M.; Gutierrez, J.; Lombardi-Hill, D.; Kamel, H.; Kernan, W.N.; Kittner, S.J.; Leira, E.C.; et al. 2021 Guideline for the Prevention of Stroke in Patients with Stroke and Transient Ischemic Attack: A Guideline From the American Heart Association/American Stroke Association. Stroke 2021, 52, E364–E467. [Google Scholar] [CrossRef]
  92. Amarenco, P.; Denison, H.; Evans, S.R.; Himmelmann, A.; James, S.; Knutsson, M.; Ladenvall, P.; Molina, C.A.; Wang, Y.; Johnston, S.C.; et al. Ticagrelor Added to Aspirin in Acute Ischemic Stroke or Transient Ischemic Attack in Prevention of Disabling Stroke: A Randomized Clinical Trial. JAMA Neurol. 2021, 78, 177–185. [Google Scholar] [CrossRef]
  93. Sahara, N.; Kuwashiro, T.; Okada, Y. Cerebral infarction and transient ischemic attack. Nihon Rinsho 2016, 74, 666–670. [Google Scholar]
  94. Cave, B.; Rawal, A.; Ardeshna, D.; Ibebuogu, U.N.; Sai-Sudhakar, C.B.; Khouzam, R.N. Targeting ticagrelor: A novel therapy for emergency reversal. Ann. Transl. Med. 2019, 7, 410. [Google Scholar] [CrossRef]
  95. Sang, Y.; Roest, M.; de Laat, B.; de Groot, P.G.; Huskens, D. Interplay between platelets and coagulation. Blood Rev. 2021, 46, 100733. [Google Scholar] [CrossRef]
  96. Zheng, B.; Li, J.; Jiang, J.; Xiang, D.; Chen, Y.; Yu, Z.; Zeng, H.; Ge, J.; Dai, X.; Liu, J.; et al. Safety and efficacy of a platelet glycoprotein Ib inhibitor for patients with non-ST segment elevation myocardial infarction: A phase Ib/IIa study. Pharmacotherapy 2021, 41, 828–836. [Google Scholar] [CrossRef]
  97. Li, T.-T.; Fan, M.-L.; Hou, S.-X.; Li, X.-Y.; Barry, D.M.; Jin, H.; Luo, S.-Y.; Kong, F.; Lau, L.-F.; Dai, X.-R.; et al. A novel snake venom-derived GPIb antagonist, anfibatide, protects mice from acute experimental ischaemic stroke and reperfusion injury. Br. J. Pharmacol. 2015, 172, 3904–3916. [Google Scholar] [CrossRef] [Green Version]
  98. Gong, P.; Li, R.; Jia, H.-Y.; Ma, Z.; Li, X.Y.; Dai, X.-R.; Luo, S.-Y. Anfibatide Preserves Blood-Brain Barrier Integrity by Inhibiting TLR4/RhoA/ROCK Pathway After Cerebral Ischemia/Reperfusion Injury in Rat. J. Mol. Neurosci. 2020, 70, 71–83. [Google Scholar] [CrossRef]
  99. Sun, Y.; Langer, H.F. Platelets, Thromboinflammation and Neurovascular Disease. Front. Immunol. 2022, 13, 843404. [Google Scholar] [CrossRef]
  100. Bartunek, J.; Barbato, E.; Heyndrickx, G.; Vanderheyden, M.; Wijns, W.; Holz, J.B. Novel antiplatelet agents: ALX-0081, a Nanobody directed towards von Willebrand factor. J. Cardiovasc. Transl. Res. 2013, 6, 355–363. [Google Scholar] [CrossRef]
  101. Kovacevic, K.D.; Jilma, B.; Zhu, S.; Gilbert, J.C.; Winter, M.-P.; Toma, A.; Hengstenberg, C.; Lang, I.; Kubica, J.; Siller-Matula, J.M. von Willebrand Factor Predicts Mortality in ACS Patients Treated with Potent P2Y12 Antagonists and is Inhibited by Aptamer BT200 Ex Vivo. Thromb. Haemost. 2020, 120, 1282–1290. [Google Scholar] [CrossRef]
  102. Kovacevic, K.D.; Greisenegger, S.; Langer, A.; Gelbenegger, G.; Buchtele, N.; Pabinger, I.; Petroczi, K.; Zhu, S.; Gilbert, J.C.; Jilma, B. The aptamer BT200 blocks von Willebrand factor and platelet function in blood of stroke patients. Sci. Rep. 2021, 11, 3092. [Google Scholar] [CrossRef]
  103. Nayak, M.K.; Son, D.J.; Fuentes, E. Modulation of Glycoprotein VI and Its Downstream Signaling Pathways as an Antiplatelet Target. Int. J. Mol. Sci. 2022, 23, 9882. [Google Scholar]
  104. Vilahur, G.; Gutiérrez, M.; Arzanauskaite, M.; Mendieta, G.; Ben-Aicha, S.; Badimon, L. Intracellular platelet signalling as a target for drug development. Vasc. Pharmacol. 2018, 111, 22–25. [Google Scholar] [CrossRef] [PubMed]
  105. Borst, O.; Gawaz, M. Glycoprotein VI—Novel target in antiplatelet medication. Pharmacol. Ther. 2021, 217, 107630. [Google Scholar] [CrossRef]
  106. Nurden, A.T. Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI. Blood Rev. 2019, 38, 100592. [Google Scholar] [CrossRef] [PubMed]
  107. Arthur, J.F.; Dunkley, S.; Andrews, R.K. Platelet glycoprotein VI-related clinical defects. Br. J. Haematol. 2007, 139, 363–372. [Google Scholar] [CrossRef] [PubMed]
  108. Dalby, A.; Mezzano, D.; Rivera, J.; Watson, S.P.; Morgan, N.V. Introduction of an ancient founder glycoprotein VI (GP6) mutation into the Chilean population. Blood Adv. 2022, 6, 5866–5869. [Google Scholar] [CrossRef]
  109. Arai, M.; Yamamoto, N.; Moroi, M.; Akamatsu, N.; Fukutake, K.; Tanoue, K. Platelets with 10% of the normal amount of glycoprotein VI have an impaired response to collagen that results in a mild bleeding tendency. Br. J. Haematol. 1995, 89, 124–130. [Google Scholar] [CrossRef]
  110. Lockyer, S.; Okuyama, K.; Begum, S.; Le, S.; Sun, B.; Watanabe, T.; Matsumoto, Y.; Yoshitake, M.; Kambayashi, J.; Tandon, N.N. GPVI-deficient mice lack collagen responses and are protected against experimentally induced pulmonary thromboembolism. Thromb. Res. 2006, 118, 371–380. [Google Scholar] [CrossRef]
  111. Ungerer, M.; Rosport, K.; Bültmann, A.; Piechatzek, R.; Uhland, K.; Schlieper, P.; Gawaz, M.; Münch, G. Novel antiplatelet drug revacept (Dimeric Glycoprotein VI-Fc) specifically and efficiently inhibited collagen-induced platelet aggregation without affecting general hemostasis in humans. Circulation 2011, 123, 1891–1899. [Google Scholar] [CrossRef]
  112. Mayer, K.; Hein-Rothweiler, R.; Schüpke, S.; Janisch, M.; Bernlochner, I.; Ndrepepa, G.; Sibbing, D.; Gori, T.; Borst, O.; Holdenrieder, S.; et al. Efficacy and Safety of Revacept, a Novel Lesion-Directed Competitive Antagonist to Platelet Glycoprotein VI, in Patients Undergoing Elective Percutaneous Coronary Intervention for Stable Ischemic Heart Disease: The Randomized, Double-blind, Placebo-Controlled ISAR-PLASTER Phase 2 Trial. JAMA Cardiol. 2021, 6, 753–761. [Google Scholar]
  113. Lebozec, K.; Jandrot-Perrus, M.; Avenard, G.; Favre-Bulle, O.; Billiald, P. Design, development and characterization of ACT017, a humanized Fab that blocks platelet’s glycoprotein VI function without causing bleeding risks. MAbs 2017, 9, 945–958. [Google Scholar] [CrossRef] [Green Version]
  114. Renaud, L.; Lebozec, K.; Voors-Pette, C.; Dogterom, P.; Billiald, P.; Perrus, M.J.; Pletan, Y.; Machacek, M. Population Pharmacokinetic/Pharmacodynamic Modeling of Glenzocimab (ACT017) a Glycoprotein VI Inhibitor of Collagen-Induced Platelet Aggregation. Pharmacomet. J. Clin. Pharmacol. 2020, 2020, 1198–1208. [Google Scholar] [CrossRef]
  115. Voors-Pette, C.; Lebozec, K.; Dogterom, P.; Jullien, L.; Billiald, P.; Ferlan, P.; Renaud, L.; Favre-Bulle, O.; Avenard, G.; Machacek, M.; et al. Safety and Tolerability, Pharmacokinetics, and Pharmacodynamics of ACT017, an Antiplatelet GPVI (Glycoprotein VI) Fab. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 956–964. [Google Scholar] [CrossRef]
  116. Sakai, K.; Someya, T.; Harada, K.; Yagi, H.; Matsui, T.; Matsumoto, M. Novel aptamer to von Willebrand factor A1 domain (TAGX-0004) shows total inhibition of thrombus formation superior to ARC1779 and comparable to caplacizumab. Haematologica 2020, 105, 2631–2638. [Google Scholar] [CrossRef] [Green Version]
  117. Markus, H.S.; McCollum, C.; Imray, C.; Goulder, M.A.; Gilbert, J.; King, A. The von Willebrand inhibitor ARC1779 reduces cerebral embolization after carotid endarterectomy: A randomized trial. Stroke 2011, 42, 2149–2153. [Google Scholar] [CrossRef] [Green Version]
  118. Lapchak, P.A.; Doyan, S.; Fan, X.; Woods, C.M. Synergistic Effect of AJW200, a von Willebrand Factor Neutralizing Antibody with Low Dose (0.9 mg/mg) Thrombolytic Therapy Following Embolic Stroke in Rabbits. J. Neurol. Neurophysiol. 2013, 4, 10.4172/2155-9562.10001466. [Google Scholar] [CrossRef]
  119. Kageyama, S.; Yamamoto, H.; Nakazawa, H.; Matsushita, J.; Kouyama, T.; Gonsho, A.; Ikeda, Y.; Yoshimoto, R. Pharmacokinetics and pharmacodynamics of AJW200, a humanized monoclonal antibody to von Willebrand factor, in monkeys. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 187–192. [Google Scholar] [CrossRef]
  120. Wu, D.; Vanhoorelbeke, K.; Cauwenberghs, N.; Meiring, M.; Depraetere, H.; Kotze, H.F.; Deckmyn, H. Inhibition of the von Willebrand (VWF)-collagen interaction by an antihuman VWF monoclonal antibody results in abolition of in vivo arterial platelet thrombus formation in baboons. Blood 2002, 99, 3623–3628. [Google Scholar] [CrossRef] [Green Version]
  121. Scully, M.; Cataland, S.R.; Peyvandi, F.; Coppo, P.; Knöbl, P.; Kremer Hovinga, J.A.; Metjian, A.; de la Rubia, J.; Pavenski, K.; Callewaert, F.; et al. Caplacizumab Treatment for Acquired Thrombotic Thrombocytopenic Purpura. N. Engl. J. Med. 2019, 380, 335–346. [Google Scholar] [CrossRef]
  122. Fontayne, A.; Meiring, M.; Lamprecht, S.; Roodt, J.; Demarsin, E.; Barbeaux, P.; Deckmyn, H. The humanized anti-glycoprotein Ib monoclonal antibody h6B4-Fab is a potent and safe antithrombotic in a high shear arterial thrombosis model in baboons. Thromb. Haemost. 2008, 100, 670–677. [Google Scholar] [CrossRef]
  123. Yang, J.; Ji, S.; Dong, N.; Zhao, Y.; Ruan, C. Engineering and characterization of a chimeric anti-platelet glycoprotein Iba monoclonal antibody and preparation of its Fab fragment. Hybridoma 2010, 29, 125–132. [Google Scholar] [CrossRef]
  124. Schulte, V.; Reusch, H.P.; Pozgajová, M.; Varga-Szabó, D.; Gachet, C.; Nieswandt, B. Two-phase antithrombotic protection after anti-glycoprotein VI treatment in mice. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 1640–1647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Nieswandt, B.; Schulte, V.; Bergmeier, W.; Mokhtari-Nejad, R.; Rackebrandt, K.; Cazenave, J.-P.; Ohlmann, P.; Gachet, C.; Zirngibl, H. Long-term antithrombotic protection by in vivo depletion of platelet glycoprotein VI in mice. J. Exp. Med. 2001, 193, 459–469. [Google Scholar] [CrossRef] [PubMed]
  126. Lin, Y.-C.; Ko, Y.-C.; Hung, S.-C.; Lin, Y.-T.; Lee, J.-H.; Tsai, J.-Y.; Kung, P.-H.; Tsai, M.-C.; Chen, Y.-F.; Wu, C.-C. Selective Inhibition of PAR4 (Protease-Activated Receptor 4)-Mediated Platelet Activation by a Synthetic Nonanticoagulant Heparin Analog. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 694–703. [Google Scholar] [CrossRef] [Green Version]
  127. Kung, P.H.; Hsieh, P.W.; Lin, Y.T.; Lee, J.H.; Chen, I.H.; Wu, C.C. HPW-RX40 prevents human platelet activation by attenuating cell surface protein disulfide isomerases. Redox Biol. 2017, 13, 266–277. [Google Scholar] [CrossRef] [PubMed]
  128. Khodier, C.; VerPlank, L.; Nag, P.P.; Pu, J.; Wurst, J.; Pilyugina, T.; Dockendorff, C.; Galinski, C.N.; Scalise, A.A.; Passam, F.; et al. Identification of ML359 as a small molecule inhibitor of protein disulfide isomerase. In Probe Reports from the NIH Molecular Libraries Program; National Center for Biotechnology Information (US): Bethesda, MD, USA, 2010. [Google Scholar]
  129. Adili, R.; Tourdot, B.E.; Mast, K.; Yeung, J.; Freedman, J.C.; Green, A.; Luci, D.K.; Jadhav, A.; Simeonov, A.; Maloney, D.J.; et al. First Selective 12-LOX Inhibitor, ML355, Impairs Thrombus Formation and Vessel Occlusion In Vivo With Minimal Effects on Hemostasis. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1828–1839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Zheng, Z.; Pinson, J.-A.; Mountford, S.J.; Orive, S.; Schoenwaelder, S.M.; Shackleford, D.; Powell, A.; Nelson, E.M.; Hamilton, J.R.; Jackson, S.P.; et al. Discovery and antiplatelet activity of a selective PI3Kβ inhibitor (MIPS-9922). Eur. J. Med. Chem. 2016, 122, 339–351. [Google Scholar] [CrossRef]
  131. Schwarz, M.; Meade, G.; Stoll, P.; Ylanne, J.; Bassler, N.; Chen, Y.C.; Hagemeyer, C.E.; Ahrens, I.; Moran, N.; Kenny, D.; et al. Conformation-specific blockade of the integrin GPIIb/IIIa: A novel antiplatelet strategy that selectively targets activated platelets. Circ. Res. 2006, 99, 25–33. [Google Scholar] [CrossRef] [Green Version]
  132. Pang, A.; Cheng, N.; Cui, Y.; Bai, Y.; Hong, Z.; Delaney, M.K.; Zhang, Y.; Chang, C.; Wang, C.; Liu, C.; et al. High-loading Gα 13-binding EXE peptide nanoparticles prevent thrombosis and protect mice from cardiac ischemia/reperfusion injury. Sci. Transl. Med. 2020, 12, eaaz7287. [Google Scholar] [CrossRef]
  133. Boldron, C.; Besse, A.; Bordes, M.F.; Tissandié, S.; Yvon, X.; Gau, B.; Badorc, A.; Rousseaux, T.; Barré, G.; Meneyrol, J.; et al. N-[6-(4-butanoyl-5-methyl-1H-pyrazol-1-yl)pyridazin-3-yl]-5-chloro-1-[2-(4-methylpiperazin-1-yl)-2-oxoethyl]-1H -indole-3-carboxamide (SAR216471), a novel intravenous and oral, reversible, and directly acting P2Y12 antagonist. J. Med. Chem. 2014, 57, 7293–7316. [Google Scholar] [CrossRef]
  134. Kong, D.; Xue, T.; Guo, B.; Cheng, J.; Liu, S.; Wei, J.; Lu, Z.; Liu, H.; Gong, G.; Lan, T.; et al. Optimization of P2Y 12 Antagonist Ethyl 6-(4-((Benzylsulfonyl)carbamoyl)piperidin-1-yl)-5-cyano-2-methylnicotinate (AZD1283) Led to the Discovery of an Oral Antiplatelet Agent with Improved Druglike Properties. J. Med. Chem. 2019, 62, 3088–3106. [Google Scholar] [CrossRef]
  135. Yang, W.; Wang, Y.; Lai, A.; Qiao, J.X.; Wang, T.C.; Hua, J.; Price, L.A.; Shen, H.; Chen, X.-Q.; Wong, P.; et al. Discovery of 4-aryl-7-hydroxyindoline based P2Y1 antagonists as novel antiplatelet agents. J. Med. Chem. 2014, 57, 6150–6164. [Google Scholar] [CrossRef]
  136. Wong, P.C.; Watson, C.; Crain, E.J. The P2Y1 receptor antagonist MRS2500 prevents carotid artery thrombosis in cynomolgus monkeys. J. Thromb. Thrombolysis 2016, 41, 514–521. [Google Scholar] [CrossRef]
  137. Gremmel, T.; Yanachkov, I.B.; Yanachkova, M.I.; Wright, G.E.; Wider, J.; Undyala, V.V.; Michelson, A.D.; Frelinger, A.L., III; Przyklenk, K. Synergistic inhibition of both P2Y1 and P2Y12 adenosine diphosphate receptors as novel approach to rapidly attenuate platelet-mediated thrombosis. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 501–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Chang, C.-H.; Chung, C.-H.; Tu, Y.-S.; Tsai, C.-C.; Hsu, C.-C.; Peng, H.-C.; Tseng, Y.J.; Huang, T.-F. Trowaglerix Venom Polypeptides as a Novel Antithrombotic Agent by Targeting Immunoglobulin-Like Domains of Glycoprotein VI in Platelet. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1307–1314. [Google Scholar] [CrossRef] [Green Version]
  139. van Eeuwijk, J.M.; Stegner, D.; Lamb, D.J.; Kraft, P.; Beck, S.; Thielmann, I.; Kiefer, F.; Walzog, B.; Stoll, G.; Nieswandt, B. The Novel Oral Syk Inhibitor, Bl1002494, Protects Mice from Arterial Thrombosis and Thromboinflammatory Brain Infarction. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1247–1253. [Google Scholar] [CrossRef] [Green Version]
  140. Tullemans, B.M.E.; Nagy, M.; Sabrkhany, S.; Griffioen, A.W.; Oude Egbrink, M.G.A.; Aarts, M.; Heemskerk, J.W.M.; Kuijpers, M.J.E. Tyrosine Kinase Inhibitor Pazopanib Inhibits Platelet Procoagulant Activity in Renal Cell Carcinoma Patients. Front. Cardiovasc. Med. 2018, 5, 142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Rigg, R.A.; Aslan, J.E.; Healy, L.D.; Wallisch, M.; Thierheimer, M.L.; Loren, C.P.; Pang, J.; Hinds, M.T.; Gruber, A.; McCarty, O.J. Oral administration of Bruton’s tyrosine kinase inhibitors impairs GPVI-mediated platelet function. Am. J. Physiol. Cell Physiol. 2016, 310, C373–C380. [Google Scholar] [CrossRef] [Green Version]
  142. Perrella, G.; Montague, S.J.; Brown, H.C.; Garcia Quintanilla, L.; Slater, A.; Stegner, D.; Thomas, M.; Heemskerk, J.W.M.; Watson, S.P. Role of Tyrosine Kinase Syk in Thrombus Stabilisation at High Shear. Int. J. Mol. Sci. 2022, 23, 493. [Google Scholar] [CrossRef]
  143. Harbi, M.H.; Smith, C.W.; Nicolson, P.L.R.; Watson, S.P.; Thomas, M.R. Novel antiplatelet strategies targeting GPVI, CLEC-2 and tyrosine kinases. Platelets 2021, 32, 29–41. [Google Scholar] [CrossRef]
  144. Nicolson, P.L.R.; Nock, S.H.; Hinds, J.; Garcia-Quintanilla, L.; Smith, C.W.; Campos, J.; Brill, A.; Pike, J.A.; Khan, A.O.; Poulter, N.S.; et al. Low-dose Btk inhibitors selectively block platelet activation by CLEC-2. Haematologica 2021, 106, 208–219. [Google Scholar] [CrossRef] [Green Version]
  145. De Candia, E. Mechanisms of platelet activation by thrombin: A short history. Thromb. Res. 2012, 129, 250–256. [Google Scholar] [CrossRef] [PubMed]
  146. Bohula, E.A.; Aylward, P.E.; Bonaca, M.P.; Corbalan, R.L.; Kiss, R.G.; Murphy, S.A.; Scirica, B.M.; White, H.; Braunwald, E.; Morrow, D.A. Efficacy and Safety of Vorapaxar with and without a Thienopyridine for Secondary Prevention in Patients with Previous Myocardial Infarction and No History of Stroke or Transient Ischemic Attack: Results from TRA 2°P-TIMI 50. Circulation 2015, 132, 1871–1879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Jones, W.S.; Tricoci, P.; Huang, Z.; Moliterno, D.J.; Harrington, R.A.; Sinnaeve, P.R.; Strony, J.; Van de Werf, F.; White, H.D.; Held, C.; et al. Vorapaxar in patients with peripheral artery disease and acute coronary syndrome: Insights from Thrombin Receptor Antagonist for Clinical Event Reduction in Acute Coronary Syndrome (TRACER). Am. Heart J. 2014, 168, 588–596. [Google Scholar] [CrossRef] [PubMed]
  148. Kosova, E.C.; Bonaca, M.P.; Dellborg, M.; He, P.; Morais, J.; Ophuis, T.O.; Scirica, B.M.; Tendera, M.; Theroux, P.; Braunwald, E.; et al. Vorapaxar in patients with coronary artery bypass grafting: Findings from the TRA 2°P-TIMI 50 trial. Eur. Heart J. Acute Cardiovasc. Care 2017, 6, 164–172. [Google Scholar] [CrossRef] [PubMed]
  149. Cavender, M.A.; Scirica, B.M.; Bonaca, M.P.; Angiolillo, D.J.; Dalby, A.J.; Dellborg, M.; Morais, J.; Murphy, S.A.; Ophuis, T.O.; Tendera, M.; et al. Vorapaxar in patients with diabetes mellitus and previous myocardial infarction: Findings from the thrombin receptor antagonist in secondary prevention of atherothrombotic ischemic events-TIMI 50 trial. Circulation 2015, 131, 1047–1053. [Google Scholar] [CrossRef] [Green Version]
  150. Franchi, F.; Rollini, F.; Faz, G.; Rivas, J.R.; Rivas, A.; Agarwal, M.; Briceno, M.; Wali, M.; Nawaz, A.; Silva, G.; et al. Pharmacodynamic Effects of Vorapaxar in Prior Myocardial Infarction Patients Treated with Potent Oral P2Y 12 Receptor Inhibitors with and Without Aspirin: Results of the VORA-PRATIC Study. J. Am. Heart Assoc. 2020, 9, e015865. [Google Scholar] [CrossRef]
  151. Gurbel, P.A.; Bliden, K.P.; Turner, S.E.; Tantry, U.S.; Gesheff, M.G.; Barr, T.P.; Covic, L.; Kuliopulos, A. Cell-Penetrating Pepducin Therapy Targeting PAR1 in Subjects with Coronary Artery Disease. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 189–197. [Google Scholar] [CrossRef] [Green Version]
  152. Kuliopulos, A.; Gurbel, P.A.; Rade, J.J.; Kimmelstiel, C.D.; Turner, S.E.; Bliden, K.P.; Fletcher, E.K.; Cox, D.H.; Covic, L.; TRIP-PCI Investigators. PAR1 (Protease-Activated Receptor 1) Pepducin Therapy Targeting Myocardial Necrosis in Coronary Artery Disease and Acute Coronary Syndrome Patients Undergoing Cardiac Catheterization: A Randomized, Placebo-Controlled, Phase 2 Study. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2990–3003. [Google Scholar] [CrossRef]
  153. Meah, M.N.; Raftis, J.; Wilson, S.J.; Perera, V.; Garonzik, S.M.; Murthy, B.; Everlof, J.G.; Aronson, R.; Luettgen, J.; Newby, D.E. Antithrombotic Effects of Combined PAR (Protease-Activated Receptor)-4 Antagonism and Factor Xa Inhibition. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2678–2685. [Google Scholar] [CrossRef]
  154. Merali, S.; Wang, Z.; Frost, C.; Callejo, M.; Hedrick, M.; Hui, L.; Shropshire, S.M.; Xu, K.; Bouvier, M.; DeSouza, M.M.; et al. New oral protease-activated receptor 4 antagonist BMS-986120: Tolerability, pharmacokinetics, pharmacodynamics, and gene variant effects in humans. Platelets 2022, 33, 969–978. [Google Scholar] [CrossRef]
  155. Wong, P.C.; Seiffert, D.; Bird, J.E.; Watson, C.A.; Bostwick, J.S.; Giancarli, M.; Allegretto, N.; Hua, J.; Harden, D.; Guay, J.; et al. Blockade of protease-activated receptor- 4(PAR4) provides robust antithrombotic activity with low bleeding. Sci. Transl. Med. 2017, 9, eaaf5294. [Google Scholar] [CrossRef]
  156. Wilson, S.J.; Ismat, F.A.; Wang, Z.; Cerra, M.; Narayan, H.; Raftis, J.; Gray, T.J.; Connell, S.; Garonzik, S.; Ma, X.; et al. PAR4 (Protease-Activated Receptor 4) Antagonism With BMS-986120 Inhibits Human Ex Vivo Thrombus Formation. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 448–456. [Google Scholar] [CrossRef] [Green Version]
  157. Nylander, S.; Kull, B.; Björkman, J.A.; Ulvinge, J.C.; Oakes, N.; Emanuelsson, B.M.; Andersson, M.; Skärby, T.; Inghardt, T.; Fjellström, O.; et al. Human target validation of phosphoinositide 3-kinase (PI3K)β: Effects on platelets and insulin sensitivity, using AZD6482 a novel PI3Kβ inhibitor. J. Thromb. Haemost. 2012, 10, 2127–2136. [Google Scholar] [CrossRef]
  158. Nylander, S.; Wågberg, F.; Andersson, M.; Skärby, T.; Gustafsson, D. Exploration of efficacy and bleeding with combined phosphoinositide 3-kinase β inhibition and aspirin in man. J. Thromb. Haemost. 2015, 13, 1494–1502. [Google Scholar] [CrossRef]
  159. Milluzzo, R.P.; Franchina, G.A.; Capodanno, D.; Angiolillo, D.J. Selatogrel, a novel P2Y 12 inhibitor: A review of the pharmacology and clinical development. Expert Opin. Investig. Drugs 2020, 29, 537–546. [Google Scholar] [CrossRef]
  160. Crescence, L.; Darbousset, R.; Caroff, E.; Hubler, F.; Riederer, M.A.; Panicot-Dubois, L.; Dubois, C. Selatogrel, a reversible P2Y12 receptor antagonist, has reduced off-target interference with haemostatic factors in a mouse thrombosis model. Thromb. Res. 2021, 200, 133–140. [Google Scholar] [CrossRef]
  161. Hardy, A.R.; Jones, M.L.; Mundell, S.J.; Poole, A.W. Reciprocal cross-talk between P2Y1 and P2Y12 receptors at the level of calcium signaling in human platelets. Blood 2004, 104, 1745–1752. [Google Scholar] [CrossRef]
  162. Tscharre, M.; Michelson, A.D.; Gremmel, T. Novel Antiplatelet Agents in Cardiovascular Disease. J. Cardiovasc. Pharmacol. Ther. 2020, 25, 191–200. [Google Scholar] [CrossRef]
  163. Kereiakes, D.J.; Henry, T.D.; DeMaria, A.N.; Bentur, O.; Carlson, M.; Seng Yue, C.; Martin, L.H.; Midkiff, J.; Mueller, M.; Meek, T.; et al. 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. [Google Scholar] [CrossRef]
  164. Bor, W.L.; Zheng, K.L.; Tavenier, A.H.; Gibson, C.M.; Granger, C.B.; Bentur, O.; Lobatto, R.; Postma, S.; Coller, B.S.; van‘t Hof, A.W.J.; et al. Pharmacokinetics, pharmacodynamics, and tolerability of subcutaneous administration of a novel glycoprotein IIb/IIIa inhibitor, RUC-4, in patients with ST-segment elevation myocardial infarction. EuroIntervention 2021, 17, 401–410. [Google Scholar] [CrossRef]
  165. Kim, K.; Hahm, E.; Li, J.; Holbrook, L.-M.; Sasikumar, P.; Stanley, R.G.; Ushio-Fukai, M.; Gibbins, J.; Cho, J. Platelet protein disulfide isomerase is required for thrombus formation but not for hemostasis in mice. Blood 2013, 122, 1052–1061. [Google Scholar] [CrossRef] [PubMed]
  166. Flaumenhaft, R.; Furie, B.; Zwicker, J.I. Therapeutic implications of protein disulfide isomerase inhibition in thrombotic disease. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 16–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Alenazy, F.O.; Thomas, M.R. Novel antiplatelet targets in the treatment of acute coronary syndromes. Platelets 2021, 32, 15–28. [Google Scholar] [CrossRef]
  168. Zwicker, J.I.; Schlechter, B.L.; Stopa, J.D.; Liebman, H.A.; Aggarwal, A.; Puligandla, M.; Caughey, T.; Bauer, K.A.; Kuemmerle, N.; Wong, E.; et al. Targeting protein disulfide isomerase with the flavonoid isoquercetin to improve hypercoagulability in advanced cancer. JCI Insight 2019, 4, e125851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Stopa, J.D.; Neuberg, D.; Puligandla, M.; Furie, B.; Flaumenhaft, R.; Zwicker, J.I. Protein disulfide isomerase inhibition blocks thrombin generation in humans by interfering with platelet factor V activation. JCI Insight 2017, 2, e89373. [Google Scholar] [CrossRef] [Green Version]
  170. Stainer, A.R.; Sasikumar, P.; Bye, A.P.; Unsworth, A.J.; Holbrook, L.M.; Tindall, M.; Lovegrove, J.A.; Gibbins, J.M. The Metabolites of the Dietary Flavonoid Quercetin Possess Potent Antithrombotic Activity, and Interact with Aspirin to Enhance Antiplatelet Effects. TH Open 2019, 3, e244–e258. [Google Scholar] [CrossRef]
  171. Ibanez, B.; Vilahur, G.; Badimon, J.J. Plaque progression and regression in atherothrombosis. J. Thromb. Haemost. 2007, 5, 292–299. [Google Scholar] [CrossRef]
  172. Badimon, L.; Bugiardini, R.; Cenko, E.; Cubedo, J.; Dorobantu, M.; Duncker, D.J.; Estruch, R.; Milicic, D.; Tousoulis, D.; Vasiljevic, Z.; et al. Position paper of the European Society of Cardiology-working group of coronary pathophysiology and microcirculation: Obesity and heart disease. Eur. Heart J. 2017, 38, 1951–1958. [Google Scholar] [CrossRef]
  173. Choi, B.; Vilahur, G.; Yadegar, D.; Viles-Gonzalez, J.; Badimon, J. The role of high-density lipoprotein cholesterol in the prevention and possible treatment of cardiovascular diseases. Curr. Mol. Med. 2006, 6, 571–587. [Google Scholar] [CrossRef]
Figure 1. Antiplatelet drugs currently used to treat ischemic heart disease and ischemic stroke. ENT: equilibrative nucleoside transporter; ASA: acetylsalicylic acid; TXA2: thromboxane A2; VWF: Von Willebrand Factor; PDR: phosphodiesterase; COX: cyclooxygenase. Figure created with BioRender.com.
Figure 1. Antiplatelet drugs currently used to treat ischemic heart disease and ischemic stroke. ENT: equilibrative nucleoside transporter; ASA: acetylsalicylic acid; TXA2: thromboxane A2; VWF: Von Willebrand Factor; PDR: phosphodiesterase; COX: cyclooxygenase. Figure created with BioRender.com.
Jcdd 09 00397 g001
Figure 3. Emerging antiplatelet targets and drugs. PDI: phosphodiesterase; VWF: Von Willebrand factor; GP: glycoprotein; PAR: protease activator receptor.
Figure 3. Emerging antiplatelet targets and drugs. PDI: phosphodiesterase; VWF: Von Willebrand factor; GP: glycoprotein; PAR: protease activator receptor.
Jcdd 09 00397 g003
Table 1. Anticoagulant reversal agents in clinical use and preclinical/clinical development.
Table 1. Anticoagulant reversal agents in clinical use and preclinical/clinical development.
In Clinical Use [13,14,15]
AgentTarget
Vitamin KWarfarin, acenocumarol
IdarucimabDabigatran
Andexanet alfaApixaban, rivaroxavan, edoxaban
Protamine sulfateUnfractionated heparin LMWH (partially)
Prothrombin complex concentrate, fresh frozen plasmaNon-specific prohemostatic agents
Preclinical/Clinical Development
AgentTarget
Aripazine (ciraparantag/PER977) (NCT04593784) [16,17]LMWH, fondaparinux, FXa inhibitors, dabigatran
γ-thrombine S195A [18]Dabigatran
GDFXa-α2M complex [19]Rivaroxaban, apixaban, dabigatran and heparins
Table 2. Factor XIIa inhibitors currently under development. This table includes the emerging FXIIa inhibitors and details the studies conducted so far to assess their efficacy and safety.
Table 2. Factor XIIa inhibitors currently under development. This table includes the emerging FXIIa inhibitors and details the studies conducted so far to assess their efficacy and safety.
Factor XIIa InhibitorsTypePhaseStudies Conducted So Far
Garadacimab (subcutaneous)AntibodyIIITested in patients with C1-esterase inhibitor-deficient hereditary angioedema showing a significant reduction of angioedema attacks. A dose-dependent increase in aPTT with no change in prothrombin time was also observed without increasing of bleeding events [34,35]. Currently ongoing phase III trials (NCT04656418, NCT04739059).
3F7 (intravenous)AntibodyPreclinicalThromboprotection in ECMO without impairing the hemostatic capacity or increasing bleeding [36,37].
9A2 and 15H8
(intravenous)
AntibodyIBoth antibodies have been shown to protect against ferric chloride-induced arterial thrombosis. 15H8 prolonged the aPTT time in non-human primates and humans and reduced fibrin formation in collagen-coated vascular grafts inserted into arteriovenous shunts in non-human primates [38].
5C12 (intravenous)AntibodyPreclinicalThromboprotection in ECMO in non-human primates [39].
Ir-CPI (intravenous)Kunitz-type serine protease inhibitorPreclinicalIt has demonstrated antithrombotic activity in:
(1) venous and arterial in vitro thrombosis models; (2) arteriovenous shunt rabbit models; and (3) extracorporeal circuit [40,41]. It can interact with factors XIIa, XIa, and Kallikrein [42].
FXII-ASO (subcutaneous)Antisense oligonucleotidePreclinicalProlonged the time to catheter thrombotic occlusion (implanted in jugular vein) compared to control in a rabbit model of thrombosis [43].
ALN-F12 (subcutaneous)Interfering RNAPreclinicalDose-dependently reduced platelet and fibrin deposition in mice models of venous and arterial thrombosis models [44].
rHA-Infestin-4 (intravenous)Kazal-type serine protease inhibitorPreclinicalProtects against arterial and venous thrombosis in mouse and rabbit models.
Reduces infarct size and brain edema formation leading to better neurological scores and survival in a mouse model of stroke [45,46,47].
aPTT: activated partial thromboplastin time; ECMO: extracorporeal membrane oxygenation.
Table 3. Factor XIa inhibitors currently under development. This table includes the emerging FXIa inhibitors and details the studies conducted so far to assess their efficacy and safety. VTE: venous thromboembolism; AF: atrial fibrilation; aPTT: activated partial thromboplastin time; MI: myocardial infarction.
Table 3. Factor XIa inhibitors currently under development. This table includes the emerging FXIa inhibitors and details the studies conducted so far to assess their efficacy and safety. VTE: venous thromboembolism; AF: atrial fibrilation; aPTT: activated partial thromboplastin time; MI: myocardial infarction.
Factor XIa InhibitorsTypePhase Studies Conducted So Far
Osocimab (subcutaneous)AntibodyIIEffective in thromboprophylaxis in patients undergoing knee arthroplasty [54].
Abelacimab (intravenous)AntibodyIIIEffective in preventing venous thromboembolism and is associated with a low bleeding risk [55]. There are ongoing phase III trials in cancer patients to compare the effect of abelacimab relative to apixaban (NCT05171049) or dalteparin (NCT05171075) in VTE recurrence and bleeding.
AB023 (Xisomab) (intravenous)AntibodyIIEffective and secure in patients with end-stage renal disease [56]. Ongoing phase II trial to test xisomab for the prevention of catheter-associated thrombosis in patients with cancer receiving chemotherapy (NCT04465760).
14E11 (subcutaneous)AntibodyPreclinicalIn mice, 14E11 has been shown to prevent arterial occlusion induced by ferric chloride to a similar degree as that accomplished by total FXI deficiency. In baboons, it has been shown to reduce platelet-rich thrombus growth in collagen-coated grafts inserted into arteriovenous shunts [57].
FXI-175, FXI-203 (intravenous)AntibodyPreclinicalFerric chloride-induced thrombosis was reduced in mice treated with FXI-175 and FX-203 compared to placebo-treated mice. Neither antibody caused severe blood loss assessed through the tail bleeding assay [58].
Frunexian EP-7041a (intravenous)Small molecule
C19H27ClN4O4
IIEP-7041 was safe and well tolerated in healthy volunteers with rapid onset and offset of action and predictable dose-related increases of aPTT [59]. In addition, there is an ongoing trial in thromboprophylaxis in COVID-19 patients (NCT05040776).
Milvexian (BMS-986177) (oral)Small molecule
C28H23Cl2F2N9O2
IIPrevention of venous thromboembolism with low risk of bleeding (phase II) [60]. In rabbits, it has demonstrated effective antithrombotic potential with limited impact on hemostasis, even when combined with aspirin [61]. A recent phase II trial (AXIOMATIC-SSP) has shown it is safe in secondary stroke prevention [62].
Asundexian (oral)Small molecule
C26H21ClF4N6O4
II bIn patients with AF, it has shown low rates of bleeding as compared with apixaban [63]. It has also shown no increase in bleeding events in MI [64] and stroke [65] patients. New phase III clinical trials have been announced to test its efficacy in patients with AF (OCEAN-AF) and in secondary prevention of stroke
(OCEAN-STROKE).
BMS-962212 (intravenous)Small molecule
C32H28ClFN8O5
ITested in healthy subjects showing good tolerance, no signs of bleeding and significant changes in aPTT and FXI clotting activity [66].
ONO-7684 (oral)Small molecule
C23H16ClF2N9O
IIt strongly inhibited factor XI coagulation activity and increased activated partial thromboplastin time [67].
BMS-654457 (intravenous)Small molecule
C36H37N5O4)
PreclinicalIt has been shown in vitro to increase aPTT without altering prothrombin time or ADP-, arachidonic acid-, or collagen-induced platelet aggregation. In rabbit models, it has shown equivalent antithrombotic effect to that achieved by standard doses of reference anticoagulants (warfarin and dabigatran) and antiplatelet agents (clopidogrel and prasugrel) in addition to reducing bleeding time [68].
ONO-5450598 (oral)Small moleculePreclinicalIt provided a significant reduction of thrombus formation as compared to rivaroxaban in a non-human primate arteriovenous shunt model of thrombosis [69].
BMS-262084 (intravenous)Small molecule
C18H31N7O5
PreclinicalEvaluated in rabbits, where it displayed antithrombotic potential in an arteriovenous-shunt model of thrombosis, and in an electrolytic-mediated carotid arterial thrombosis [70].
FXI-ASO (ISIS416858) (subcutaneous)Antisense oligonucleotideIIEffective in thromboprophylaxis in patients undergoing knee arthroplasty [71].
Table 4. Factor IXa inhibitors currently under development. This table includes the emerging FIXa inhibitors and details the studies conducted so far to assess their efficacy and safety.
Table 4. Factor IXa inhibitors currently under development. This table includes the emerging FIXa inhibitors and details the studies conducted so far to assess their efficacy and safety.
Factor IXa InhibitorsTypePhaseStudies Conducted So Far
Pegnivacogin (intravenous)RNA aptamerII
-
Phase II trial in NSTEMI patients undergoing cardiac catheterization did not show significant differences compared with heparin [75].
-
A randomized clinical trial in patients undergoing percutaneous coronary intervention had to be terminated early due to severe allergic reactions [76].
-
It has decreased platelet activation and aggregation in vitro [77].
SB249417 (intravenous)AntibodyIIt has demonstrated prolongation of coagulation measures in humans [78].
TTP889 (oral)Small moleculeIIIt has not been shown to be effective for the extended prevention of venous thromboembolism [79].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Barriuso, I.; Worner, F.; Vilahur, G. Novel Antithrombotic Agents in Ischemic Cardiovascular Disease: Progress in the Search for the Optimal Treatment. J. Cardiovasc. Dev. Dis. 2022, 9, 397. https://doi.org/10.3390/jcdd9110397

AMA Style

Barriuso I, Worner F, Vilahur G. Novel Antithrombotic Agents in Ischemic Cardiovascular Disease: Progress in the Search for the Optimal Treatment. Journal of Cardiovascular Development and Disease. 2022; 9(11):397. https://doi.org/10.3390/jcdd9110397

Chicago/Turabian Style

Barriuso, Ignacio, Fernando Worner, and Gemma Vilahur. 2022. "Novel Antithrombotic Agents in Ischemic Cardiovascular Disease: Progress in the Search for the Optimal Treatment" Journal of Cardiovascular Development and Disease 9, no. 11: 397. https://doi.org/10.3390/jcdd9110397

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

Article Metrics

Back to TopTop