[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (107)

Search Parameters:
Keywords = NOAC

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
87 pages, 3719 KiB  
Review
Beyond Anticoagulation: A Comprehensive Review of Non-Vitamin K Oral Anticoagulants (NOACs) in Inflammation and Protease-Activated Receptor Signaling
by Shirin Jannati, Rajashree Patnaik and Yajnavalka Banerjee
Int. J. Mol. Sci. 2024, 25(16), 8727; https://doi.org/10.3390/ijms25168727 (registering DOI) - 10 Aug 2024
Viewed by 239
Abstract
Non-vitamin K oral anticoagulants (NOACs) have revolutionized anticoagulant therapy, offering improved safety and efficacy over traditional agents like warfarin. This review comprehensively examines the dual roles of NOACs—apixaban, rivaroxaban, edoxaban, and dabigatran—not only as anticoagulants, but also as modulators of inflammation via protease-activated [...] Read more.
Non-vitamin K oral anticoagulants (NOACs) have revolutionized anticoagulant therapy, offering improved safety and efficacy over traditional agents like warfarin. This review comprehensively examines the dual roles of NOACs—apixaban, rivaroxaban, edoxaban, and dabigatran—not only as anticoagulants, but also as modulators of inflammation via protease-activated receptor (PAR) signaling. We highlight the unique pharmacotherapeutic properties of each NOAC, supported by key clinical trials demonstrating their effectiveness in preventing thromboembolic events. Beyond their established anticoagulant roles, emerging research suggests that NOACs influence inflammation through PAR signaling pathways, implicating factors such as factor Xa (FXa) and thrombin in the modulation of inflammatory responses. This review synthesizes current evidence on the anti-inflammatory potential of NOACs, exploring their impact on inflammatory markers and conditions like atherosclerosis and diabetes. By delineating the mechanisms by which NOACs mediate anti-inflammatory effects, this work aims to expand their therapeutic utility, offering new perspectives for managing inflammatory diseases. Our findings underscore the broader clinical implications of NOACs, advocating for their consideration in therapeutic strategies aimed at addressing inflammation-related pathologies. This comprehensive synthesis not only enhances understanding of NOACs’ multifaceted roles, but also paves the way for future research and clinical applications in inflammation and cardiovascular health. Full article
(This article belongs to the Special Issue New Trends in Diabetes, Hypertension and Cardiovascular Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>The current model of the blood coagulation cascade, depicting NOACs’ mechanism of action. There are two pathways, the intrinsic pathway and the extrinsic pathway. These multicomponent processes are illustrated as enzymes, inhibitors, zymogens, or complexes. On injury to the vessel wall, tissue factor, the cofactor for the extrinsic tenase complex, is exposed to circulating FVIIa and forms the extrinsic tenase. FIX and FX are converted to their serine proteases FIXa and FXa, which then form the intrinsic tenase and the prothrombinase complexes, respectively. The combined actions of the intrinsic and extrinsic tenase and the prothrombinase complexes lead to an explosive burst of the enzyme thrombin (IIa). In addition to its multiple procoagulant roles, thrombin also acts in an anticoagulant capacity when combined with the cofactor thrombomodulin in the protein Case complex. The product of the protein Case reaction, activated protein C (APC), inactivates the cofactors FVa and FVIIIa. The cleaved species, FVai and FVIIIai, no longer support the respective procoagulant activities. Once thrombin is generated through procoagulant mechanisms, thrombin cleaves fibrinogen (releasing fibrinopeptide A and B [FPA and FPB]), as well as activating FXIII to form a cross-linked fibrin clot. Thrombin–thrombomodulin also activates thrombin activate-able fibrinolysis inhibitor, which slows fibrin degradation by plasmin. The procoagulant response is downregulated by the stoichiometric inhibitor tissue factor pathway inhibitor (TFPI) and antithrombin III (AT-III). TFPI serves to attenuate the activity of the extrinsic tenase trigger of coagulation. AT-III directly inhibits thrombin, FIXa, and FXa. The accessory pathway provides an alternate route for the generation of FIXa. Thrombin has also been shown to activate FXI. The fibrin clot is eventually degraded by plasmin, yielding soluble fibrin peptides. Factor Xa inhibitors (apixaban, edoxaban, and rivaroxaban) act by binding to the active site of factor Xa, inhibiting the conversion of prothrombin to thrombin, the final enzyme in the coagulation cascade. Dabigatran, conversely, functions as a direct thrombin inhibitor. It binds with high affinity to the active site of thrombin, inhibiting its ability to convert fibrinogen to fibrin, thereby preventing clot formation.</p>
Full article ">Figure 2
<p>Schematic representation of structural domains of FXa and binding sites of FXa inhibitors. A schematic representation of FXa protein structural domains and the location of the binding sites of FXa inhibitors (apixaban, edoxaban, and rivaroxaban) are depicted. (<b>A</b>) illustrates the domain organization of FXa, highlighting the serine protease domain in the heavy chain (indicated by green) where the binding site is located. (<b>B</b>–<b>D</b>) depict the structures of apixaban, edoxaban, and rivaroxaban, respectively, as obtained from the Protein Data Bank, indicating their binding sites with FXa, which exhibit enzyme kinetics similar to competitive inhibitors.</p>
Full article ">Figure 3
<p>Schematic representation of PAR1- and PAR2-mediated signal transduction. PAR1 and PAR2 are G protein-coupled receptors that can be activated by thrombin and FXa, initiating a cascade of cellular responses. Upon cleavage, PAR1/2 interact with different G proteins like Gα<sub>i</sub>, Gα<sub>12/13</sub>, Gα<sub>s</sub>, and Gα<sub>q</sub>. Gα<sub>12/13</sub> leads to Ras homolog family member A (RhoA) activation, via Rho guanine nucleotide exchange factors (RhoGEFs) influencing cell hypertrophy. Gα<sub>q</sub> activates phospholipase C-β, generating second messengers that trigger calcium release and Protein Kinase C (PKC) activation. PKC can further activate the nuclear factor kappa B (NF-κB) signaling pathway to upregulate production of SRY-box transcription factor 4 (SOX4) and A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5). Gα<sub>i</sub> can inhibit adenylate cyclase (AC) to regulate downstream cAMP, whereas Gα<sub>s</sub> can increase cAMP. β-arrestin can activate the ERK1/2 signaling pathway but exhibits inhibitory effects on PKC and calcium release. Anticoagulants like dabigatran (thrombin inhibitor) and apixaban, edoxaban, and rivaroxaban (FXa inhibitors) can potentially disrupt this signaling by preventing PAR activation. Ultimately, these signal transduction pathways can trigger physiological changes like inflammatory and immune responses, cell hypertrophy, and cell migration.</p>
Full article ">Figure 4
<p>Schematic representation of PAR4-mediated signal transduction. PAR4 can activate signaling pathways involving Gα<sub>12/13</sub> and Gα<sub>q</sub>. Gα<sub>12/13</sub> prompts RhoGEFs to activate RhoA, while Gα<sub>q</sub> -Phospholipase C- β (PLC- β) leads to downstream effects such as upregulation of inositol triphosphate (IP3) and diacylglycerol (DAG), resulting in calcium alterations and PKC upregulation, which ultimately leads to activation of the NF-κB signaling pathway. Additionally, β-arrestin can facilitate ERK1/2 phosphorylation but has inhibitory effects on PKC. Thrombin, known for its ability to cleave PAR4, can influence these pathways, therefore dabigatran (thrombin inhibitor) can modulate signal transduction by attenuating thrombin’s effects. As seen, PAR4 activation can cause physiological alterations such as inflammatory and immune response, endothelial barrier dysfunction, and platelet activation.</p>
Full article ">Figure 5
<p>Proposed mechanism of apixaban’s modulatory effects on factor Xa and associated inflammatory signaling pathways in an osteoarthritic chondrocyte model. This illustration delineates the pathways through which apixaban may exert anti-inflammation in the context of osteoarthritis. Apixaban targets FXa, inhibiting its ability to bind and activate PAR2, which is represented by the red inhibitory line. This intervention most likely attenuates the downstream signaling cascades involved in OA pathophysiology: 1. PAR2 inhibition: The blockage of PAR2 activation by apixaban may ameliorate the downstream signaling events mediated by ERK1/2 that lead to the production of pro-inflammatory cytokines, such as TNF-α and IL-1β, potentially alleviating chronic pain associated with OA. 2. Cytokine modulation: The expected reduction in TNF-α and IL-1β due to apixaban’s action on FXa mitigates the upregulation of molecules like MCP-1, which are involved in monocyte recruitment and the NF-κB signaling pathway, both key contributors to inflammation and osteoclastogenesis. 3. Protein expression: The illustration also indicates the potential effects of apixaban on the expression of regulatory proteins, including SOX4 and ADAMTS5, and their impact on critical components like aggrecan, which is essential for cartilage integrity. 4. Chondrocyte integrity and bone health: By modulating these inflammatory and catabolic pathways, apixaban may help preserve chondrocyte integrity, mitigate the generation of reactive oxygen species (ROS), and contribute to maintaining joint health by potentially impacting the RANK/RANKL pathway, which is crucial for osteoclast activity and bone resorption.</p>
Full article ">
26 pages, 894 KiB  
Review
Comparative Cardioprotective Effectiveness: NOACs vs. Nattokinase—Bridging Basic Research to Clinical Findings
by Maja Muric, Marina Nikolic, Andreja Todorovic, Vladimir Jakovljevic and Ksenija Vucicevic
Biomolecules 2024, 14(8), 956; https://doi.org/10.3390/biom14080956 - 7 Aug 2024
Viewed by 396
Abstract
The use of non-vitamin K antagonist oral anticoagulants (NOACs) has brought a significant progress in the management of cardiovascular diseases, considered clinically superior to vitamin K antagonists (VKAs) particularly in the prevention and treatment of thromboembolic events. In addition, numerous advantages such as [...] Read more.
The use of non-vitamin K antagonist oral anticoagulants (NOACs) has brought a significant progress in the management of cardiovascular diseases, considered clinically superior to vitamin K antagonists (VKAs) particularly in the prevention and treatment of thromboembolic events. In addition, numerous advantages such as fixed dosing, lack of laboratory monitoring, and fewer food and drug-to-drug interactions make the use of NOACs superior to VKAs. While NOACs are synthetic drugs prescribed for specific conditions, nattokinase (NK) is a natural enzyme derived from food that has potential health benefits. Various experimental and clinical studies reported the positive effects of NK on the circulatory system, including the thinning of blood and the dissolution of blood clots. This enzyme showed not only fibrinolytic activity due to its ability to degrade fibrin, but also an affinity as a substrate for plasmin. Recent studies have shown that NK has additional cardioprotective effects, such as antihypertensive and anti-atherosclerotic effects. In this narrative review, we presented the cardioprotective properties of two different approaches that go beyond anticoagulation: NOACs and NK. By combining evidence from basic research with clinical findings, we aim to elucidate the comparative cardioprotective efficacy of these interventions and highlight their respective roles in modern cardiovascular care. Full article
Show Figures

Figure 1

Figure 1
<p>Coagulation cascade and signaling pathways of NOACs and NK. Abbreviations: NOACs—non-vitamin K antagonist oral anticoagulants; TF—tissue factor; PL—platelet phospholipids; u-PA—urokinase-plasminogen activator; t-PA—tissue-plasminogen activator; PAI-1—Plasminogen activator inhibitor-1; PAI-2—Plasminogen activator inhibitor-2.</p>
Full article ">Figure 2
<p>Anti-atherosclerotic mechanisms of both NOACs and NK. Abbreviations: NOACs—non-vitamin K antagonist oral anticoagulants; FXa—factor Xa; u-PA—urokinase-plasminogen activator; VCAM1—vascular cell adhesion molecule-1; CCL5—CC-chemokine ligand 5; NLRP3—NLR family pyrin domain containing 3; TLR4—toll like receptor 4; PAR—protease activated receptor; TGF-β—transforming growth factor-β; TNF-α—tumor necrosis factor α; COL3A1—collagen type III, alpha-1 chain; IL—interleukin; MMP9—matrix metalloproteinase 9; MCP1—monocyte chemoattractant protein 1; ICAM1—intercellular adhesion molecule 1; eNOS—endothelial nitric oxide synthase; ANP—atrial natriuretic peptide; BNP—brain natriuretic peptide; p-ERK—phosphorylated extracellular signal-regulated kinase; ROS—reactive oxygen species; Bcl-2—the B cell lymphoma-2; t-PA—tissue-plasminogen activator; TC—total cholesterol; TG—triglycerides; LDL—low-density lipoprotein; HDL—high-density lipoprotein; JAK—janus kinase; STAT1—signal transducers and activators of transcription 1; TRAF-6—tumor necrosis factor receptor-associated factor 6; Nf-kβ—nuclear factor kappa-light-chain-enhancer of activated B cells; MAPKs—mitogen-activated protein kinases; NO—nitrite oxide; CK—creatine kinase; CK-MB—creatine kinase-myoglobin binding; LDH—lactate dehydrogenase.</p>
Full article ">
14 pages, 1090 KiB  
Article
Ex Vivo Antiplatelet Effects of Oral Anticoagulants
by Giulia Renda, Valentina Bucciarelli, Giulia Barbieri, Paola Lanuti, Martina Berteotti, Gelsomina Malatesta, Francesca Cesari, Tanya Salvatore, Betti Giusti, Anna Maria Gori, Rossella Marcucci and Raffaele De Caterina
J. Cardiovasc. Dev. Dis. 2024, 11(4), 111; https://doi.org/10.3390/jcdd11040111 - 31 Mar 2024
Viewed by 1121
Abstract
Background: The impact of non-vitamin K antagonist oral anticoagulants (NOACs) on platelet function is still unclear. We conducted a comprehensive ex vivo study aimed at assessing the effect of the four currently marketed NOACs on platelet function. Methods: We incubated blood samples from [...] Read more.
Background: The impact of non-vitamin K antagonist oral anticoagulants (NOACs) on platelet function is still unclear. We conducted a comprehensive ex vivo study aimed at assessing the effect of the four currently marketed NOACs on platelet function. Methods: We incubated blood samples from healthy donors with concentrations of NOACs (50, 150 and 250 ng/mL), in the range of those achieved in the plasma of patients during therapy. We evaluated generation of thrombin; light transmittance platelet aggregation (LTA) in response to adenosine diphosphate (ADP), thrombin receptor-activating peptide (TRAP), human γ-thrombin (THR) and tissue factor (TF); generation of thromboxane (TX)B2; and expression of protease-activated receptor (PAR)-1 and P-selectin on the platelet surface. Results: All NOACs concentration-dependently reduced thrombin generation compared with control. THR-induced LTA was suppressed by the addition of dabigatran at any concentration, while TF-induced LTA was reduced by factor-Xa inhibitors. ADP- and TRAP-induced LTA was not modified by NOACs. TXB2 generation was reduced by all NOACs, particularly at the highest concentrations. We found a concentration-dependent increase in PAR-1 expression after incubation with dabigatran, mainly at the highest concentrations, but not with FXa inhibitors; P-selectin expression was not changed by any drugs. Conclusions: Treatment with the NOACs is associated with measurable ex vivo changes in platelet function, arguing for antiplatelet effects beyond the well-known anticoagulant activities of these drugs. There are differences, however, among the NOACs, especially between dabigatran and the FXa inhibitors. Full article
Show Figures

Figure 1

Figure 1
<p>Thrombin generation parameters after the addition of increasing concentrations of dabigatran, rivaroxaban, apixaban and edoxaban. Dot plots report all single values of endogenous thrombin potential (ETP, panel (<b>A</b>)) and peak of thrombin generation (panel (<b>B</b>)). The horizontal lines represent the mean values. * <span class="html-italic">p</span> &lt; 0.0001, ° <span class="html-italic">p</span> &lt; 0.001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>§</sup> <span class="html-italic">p</span> &lt; 0.05 vs. the corresponding controls. C = control; 50 = 50 ng/mL; 150 = 150 ng/mL; 250 = 250 ng/mL.</p>
Full article ">Figure 2
<p>Platelet aggregation induced by different agents after the addition of increasing concentrations of dabigatran, rivaroxaban, apixaban and edoxaban. Box and whiskers Tukey plot reporting the median, 25th and 75th percentile and range for values of platelet aggregation in platelet-rich plasma (optical transmittance, in percent of maximum aggregation) induced by ADP (<b>A</b>), thrombin receptor-activating peptide (TRAP) (<b>B</b>), gamma-thrombin (THR) (<b>C</b>) and tissue factor (TF) (<b>D</b>). The asterisk within the bars reports the mean values. * <span class="html-italic">p</span> &lt; 0.0001, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01, vs. corresponding controls.</p>
Full article ">Figure 3
<p>Serum TXB<sub>2</sub> generation after the addition of increasing concentrations of dabigatran, rivaroxaban, apixaban and edoxaban. Box and whiskers Tukey plot reporting the median, 25th and 75th percentile and range for values of serum TXB<sub>2</sub>. The asterisk within the bars reports the mean values. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>§</sup> <span class="html-italic">p</span> &lt; 0.05 vs. corresponding control.</p>
Full article ">Figure 4
<p>Expression of PAR-1 on platelet surface using flow cytometry without and with the addition of increasing concentrations of dabigatran. Box and whiskers Tukey plot reporting the median, 25th and 75th percentile and range for values of mean fluorescence intensity (MFI) ratio. The «+» within the bars indicate the mean values. ° <span class="html-italic">p</span> &lt; 0.001 vs. control; <sup>§</sup> <span class="html-italic">p</span> &lt; 0.05 vs. control.</p>
Full article ">
17 pages, 1761 KiB  
Article
Optimizing Atrial Fibrillation Care: Comparative Assessment of Anticoagulant Therapies and Risk Factors
by Marius Rus, Adriana Ioana Ardelean, Simina Crisan, Paula Marian, Oana Lilliana Pobirci, Veronica Huplea, Claudia Judea Pusta, Gheorghe Adrian Osiceanu, Claudia Elena Stanis and Felicia Liana Andronie-Cioara
Clin. Pract. 2024, 14(1), 344-360; https://doi.org/10.3390/clinpract14010027 - 12 Feb 2024
Viewed by 1821
Abstract
Background and objectives: Atrial fibrillation (AF) is a common arrhythmia associated with various risk factors and significant morbidity and mortality. Materials and methods: This article presents findings from a study involving 345 patients with permanent AF. This study examined demographics, risk factors, associated [...] Read more.
Background and objectives: Atrial fibrillation (AF) is a common arrhythmia associated with various risk factors and significant morbidity and mortality. Materials and methods: This article presents findings from a study involving 345 patients with permanent AF. This study examined demographics, risk factors, associated pathologies, complications, and anticoagulant therapy over the course of a year. Results: The results showed a slight predominance of AF in males (55%), with the highest incidence in individuals aged 75 and older (49%). Common risk factors included arterial hypertension (54%), dyslipidemia, diabetes mellitus type 2 (19.13%), and obesity (15.65%). Comorbidities such as congestive heart failure (35.6%), mitral valve regurgitation (60%), and dilated cardiomyopathy (32%) were prevalent among the patients. Major complications included congestive heart failure (32%), stroke (17%), and myocardial infarction (5%). Thromboembolic and bleeding risk assessment using CHA2DS2-VASc and HAS-BLED scores demonstrated a high thromboembolic risk in all patients. The majority of patients were receiving novel oral anticoagulants (NOACs) before admission (73%), while NOACs were also the most prescribed antithrombotic therapy at discharge (61%). Conclusions: This study highlights the importance of risk factor management and appropriate anticoagulant therapy in patients with AF, to reduce complications and improve outcomes. The results support the importance of tailored therapeutic schemes, for optimal care of patients with AF. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Sex distribution.</p>
Full article ">Figure 2
<p>Age distribution.</p>
Full article ">Figure 3
<p>Hospitalization duration.</p>
Full article ">Figure 4
<p>Complications.</p>
Full article ">Figure 5
<p>Comorbidities.</p>
Full article ">Figure 6
<p>Antithrombotic medication before admission.</p>
Full article ">Figure 7
<p>Antithrombotic medication during hospitalization.</p>
Full article ">Figure 8
<p>Antithrombotic therapy prescribed at discharge.</p>
Full article ">
17 pages, 7284 KiB  
Article
Novel Thiourea and Oxime Ether Isosteviol-Based Anticoagulants: MD Simulation and ADMET Prediction
by Marcin Gackowski, Mateusz Jędrzejewski, Sri Satya Medicharla, Rajesh Kondabala, Burhanuddin Madriwala, Katarzyna Mądra-Gackowska and Renata Studzińska
Pharmaceuticals 2024, 17(2), 163; https://doi.org/10.3390/ph17020163 - 28 Jan 2024
Viewed by 1261
Abstract
Activated blood coagulation factor X (FXa) plays a critical initiation step of the blood-coagulation pathway and is considered a desirable target for anticoagulant drug development. It is reversibly inhibited by nonvitamin K antagonist oral anticoagulants (NOACs) such as apixaban, betrixaban, edoxaban, and rivaroxaban. [...] Read more.
Activated blood coagulation factor X (FXa) plays a critical initiation step of the blood-coagulation pathway and is considered a desirable target for anticoagulant drug development. It is reversibly inhibited by nonvitamin K antagonist oral anticoagulants (NOACs) such as apixaban, betrixaban, edoxaban, and rivaroxaban. Thrombosis is extremely common and is one of the leading causes of death in developed countries. In previous studies, novel thiourea and oxime ether isosteviol derivatives as FXa inhibitors were designed through a combination of QSAR studies and molecular docking. In the present contribution, molecular dynamics (MD) simulations were performed for 100 ns to assess binding structures previously predicted by docking and furnish additional information. Moreover, three thiourea- and six oxime ether-designed isosteviol analogs were then examined for their drug-like and ADMET properties. MD simulations demonstrated that four out of the nine investigated isosteviol derivatives, i.e., one thiourea and three oxime ether ISV analogs, form stable complexes with FXa. These derivatives interact with FXa in a manner similar to Food and Drug Administration (FDA)-approved drugs like edoxaban and betrixaban, indicating their potential to inhibit factor Xa activity. One of these derivatives, E24, displays favorable pharmacokinetic properties, positioning it as the most promising drug candidate. This, along with the other three derivatives, can undergo further chemical synthesis and bioassessment. Full article
Show Figures

Figure 1

Figure 1
<p>The crystal structure of <span class="html-italic">H. sapiens</span> FXa (red) in complex with a calcium cation (yellow) and an inhibitor; PDB code: 2P16. The inset shows the binding pocket, with key residues highlighted. The binding pocket comprises four subpockets: S1 (constructed by C191, Q192, D194, and I227), S2 (C219), S3 (E147 and G218), and S4 (Y99, F174, W215, G216, and E217).</p>
Full article ">Figure 2
<p>Structures of complexes between ISV derivatives and FXa (red) obtained through docking (<b>A</b>–<b>I</b>). These structures served as the initial configurations for the subsequent MD simulations.</p>
Full article ">Figure 3
<p>RMSD values of protein backbone (red) and ligand (blue) for the simulation of an FXa complex with different ligands (<b>A</b>–<b>I</b>). While the RMSD for the protein backbone remains stable with consistently small values across all simulations, the RMSD of the ligand varies between simulations, indicating distinct stability levels of the complexes.</p>
Full article ">Figure 4
<p>The distance between the CoM of the ligand and the CoM of FXa throughout the simulations (<b>A</b>–<b>I</b>). Typically, the distance remains around 15 to 20 Å, except for the FXa complex with E04. In this case, after 15 ns, the distance increases rapidly, stabilizing at 25 Å. This observation implies the dissociation of the ligand and its potential rebinding at a different site.</p>
Full article ">Figure 5
<p>The RMSD changes of amino acids forming the binding pocket (Y99, E147, F174, C191, Q192, D194, W215, G216, E217, G218, C219, and I227) (<b>A</b>–<b>I</b>). The RMSD for FXa complexes with E10, E20, and E21 remains relatively stable, with low values ranging from 2 to 3 Å. In contrast, other complexes show larger RMSD changes, indicating diverse mobility of the ligand-binding site, dependent on the specific type of ligand bound.</p>
Full article ">Figure 6
<p>Representative ligand conformations bound to FXa. Conformations were chosen through ligand clustering using the GROMOS algorithm with a cutoff of 1.0 Å, based on frames sampled every 100 ps. Conformations found in less than 5% of frames are not shown for clarity. (<b>A</b>): E10, (<b>B</b>): E15, (<b>C</b>): E20, and (<b>D</b>): E24.</p>
Full article ">Figure 7
<p>Snapshots from MD simulations for the most representative conformations of the ligands shown in <a href="#pharmaceuticals-17-00163-f006" class="html-fig">Figure 6</a>. The stick representation depicts the side chains of key residues and ligands. Hydrogen bonds are shown with dashed lines. (<b>A</b>): E10, (<b>B</b>): E15, (<b>C</b>): E20, and (<b>D</b>): E24.</p>
Full article ">Figure 8
<p>Chemical structures of FDA-approved FXa inhibitors.</p>
Full article ">
11 pages, 703 KiB  
Article
One-Day Interruption of NOAC Is Associated with Low Risk of Periprocedural Adverse Events during Pulmonary Vein Isolation If Combined with Left Atrial Thrombus Exclusion with Computed Tomography
by Katalin Piros, Adorján Vida, Nándor Szegedi, Péter Perge, Zoltán Salló, Arnold Béla Ferencz, Vivien Klaudia Nagy, Szilvia Herczeg, Pál Ábrahám, Csaba Csobay-Novák, Zsófia Drobni, Tamás Tahin, Györgyi Apponyi, Béla Merkely, László Gellér and István Osztheimer
Life 2024, 14(1), 133; https://doi.org/10.3390/life14010133 - 17 Jan 2024
Viewed by 963
Abstract
Background: Safety, efficacy, and patient comfort are the expectations during pulmonary vein isolation (PVI). We aimed to validate the combined advantages of pre- and periprocedural anticoagulation with non-vitamin K anticoagulants (NOACs) and rigorous left atrial appendage thrombus (LAAT) exclusion with computed tomography (CT). [...] Read more.
Background: Safety, efficacy, and patient comfort are the expectations during pulmonary vein isolation (PVI). We aimed to validate the combined advantages of pre- and periprocedural anticoagulation with non-vitamin K anticoagulants (NOACs) and rigorous left atrial appendage thrombus (LAAT) exclusion with computed tomography (CT). Methods: This study included a population of consecutive patients, between March 2018 and June 2020, who underwent cardiac CT within 24 h before PVI to guide the ablation and rule out LAAT. NOAC was omitted 24 h before the ablation. Results: A total of 187 patients (63% male) underwent CT before PVI. None of the patients experienced stroke during or after the procedure. The complication rate was low, with no thromboembolic events and 2.1% of patients experiencing a major bleeding event. Conclusions: Omitting NOAC 24 h before the ablation might be safe if combined with left atrial thrombus exclusion with computed tomography. Full article
(This article belongs to the Special Issue Feature Studies in Diagnostic Radiology)
Show Figures

Figure 1

Figure 1
<p>Workflow.</p>
Full article ">Figure 2
<p>CTA image of the heart, with a yellow arrow indicating a possible left atrial appendage thrombus, which is more pronounced in the delayed-phase images. ((<b>A</b>) no thrombus Coronary CT, (<b>B</b>) no thrombus Delayed phase, (<b>C</b>): inconclusive for thrombus coronary CT, (<b>D</b>): inconclusive for thrombus Delayed phase).</p>
Full article ">
14 pages, 2244 KiB  
Systematic Review
Non-Vitamin K Antagonist Oral Anticoagulants versus Low Molecular Weight Heparin for Cancer-Related Venous Thromboembolic Events: Individual Patient Data Meta-Analysis
by Chun En Yau, Chen Ee Low, Natasha Yixuan Ong, Sounak Rana, Lucas Jun Rong Chew, Sara Moiz Tyebally, Ping Chai, Tiong-Cheng Yeo, Mark Y. Chan, Matilda Xinwei Lee, Li-Ling Tan, Chieh-Yang Koo, Ainsley Ryan Yan Bin Lee and Ching-Hui Sia
Cancers 2023, 15(24), 5887; https://doi.org/10.3390/cancers15245887 - 18 Dec 2023
Cited by 2 | Viewed by 1471
Abstract
Venous thromboembolism (VTE) is a leading cause of morbidity and mortality in cancer patients. Low molecular weight heparin (LMWH) has been the standard of care but new guidelines have approved the use of non-vitamin K antagonist oral anticoagulants (NOAC). By conducting an individual [...] Read more.
Venous thromboembolism (VTE) is a leading cause of morbidity and mortality in cancer patients. Low molecular weight heparin (LMWH) has been the standard of care but new guidelines have approved the use of non-vitamin K antagonist oral anticoagulants (NOAC). By conducting an individual patient data (IPD) meta-analysis of randomised controlled trials (RCTs) comparing the outcomes of NOAC versus LMWH in cancer patients, we aim to determine an ideal strategy for the prophylaxis of VTE and prevention of VTE recurrence. Three databases were searched from inception until 19 October 2022. IPD was reconstructed from Kaplan–Meier curves. Shared frailty, stratified Cox and Royston–Parmar models were fit to compare the outcomes of venous thromboembolism recurrence and major bleeding. For studies without Kaplan–Meier curves, aggregate data meta-analysis was conducted using random-effects models. Eleven RCTs involving 4844 patients were included. Aggregate data meta-analysis showed that administering NOACs led to a significantly lower risk of recurrent VTE (RR = 0.65; 95%CI: 0.50–0.84) and deep vein thrombosis (DVT) (RR = 0.60; 95%CI: 0.40–0.90). In the IPD meta-analysis, NOAC when compared with LMWH has an HR of 0.65 (95%CI: 0.49–0.86) for VTE recurrence. Stratified Cox and Royston–Parmar models demonstrated similar results. In reducing risks of recurrent VTE and DVT among cancer patients, NOACs are superior to LMWHs without increased major bleeding. Full article
(This article belongs to the Special Issue Molecular Insights into Drug Resistance in Cancer)
Show Figures

Figure 1

Figure 1
<p>Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flowchart.</p>
Full article ">Figure 2
<p>Forest plot of recurrent venous thromboembolism outcomes [<a href="#B7-cancers-15-05887" class="html-bibr">7</a>,<a href="#B15-cancers-15-05887" class="html-bibr">15</a>,<a href="#B16-cancers-15-05887" class="html-bibr">16</a>,<a href="#B23-cancers-15-05887" class="html-bibr">23</a>,<a href="#B24-cancers-15-05887" class="html-bibr">24</a>,<a href="#B35-cancers-15-05887" class="html-bibr">35</a>,<a href="#B37-cancers-15-05887" class="html-bibr">37</a>]. Vertical reference line indicates a risk ratio of 1. Diamond represents the aggregated effect size.</p>
Full article ">Figure 3
<p>Forest plot of recurrent deep vein thrombosis outcomes [<a href="#B7-cancers-15-05887" class="html-bibr">7</a>,<a href="#B15-cancers-15-05887" class="html-bibr">15</a>,<a href="#B23-cancers-15-05887" class="html-bibr">23</a>,<a href="#B36-cancers-15-05887" class="html-bibr">36</a>,<a href="#B37-cancers-15-05887" class="html-bibr">37</a>]. Vertical reference line indicates a risk ratio of 1. Diamond represents the aggregated effect size.</p>
Full article ">Figure 4
<p>Forest plot of major bleeding outcomes [<a href="#B7-cancers-15-05887" class="html-bibr">7</a>,<a href="#B15-cancers-15-05887" class="html-bibr">15</a>,<a href="#B16-cancers-15-05887" class="html-bibr">16</a>,<a href="#B23-cancers-15-05887" class="html-bibr">23</a>,<a href="#B24-cancers-15-05887" class="html-bibr">24</a>,<a href="#B33-cancers-15-05887" class="html-bibr">33</a>,<a href="#B34-cancers-15-05887" class="html-bibr">34</a>,<a href="#B35-cancers-15-05887" class="html-bibr">35</a>,<a href="#B36-cancers-15-05887" class="html-bibr">36</a>,<a href="#B37-cancers-15-05887" class="html-bibr">37</a>]. Vertical reference line indicates a risk ratio of 1. Diamond represents the aggregated effect size.</p>
Full article ">Figure 5
<p>Cumulative incidence curve of pooled cohorts for recurrent venous thromboembolism.</p>
Full article ">Figure 6
<p>Cumulative incidence curve of pooled cohorts for major bleeding.</p>
Full article ">
12 pages, 1054 KiB  
Article
Prolonged Secondary Stroke Prevention with Edoxaban: A Long-Term Follow-Up of the SATES Study
by Irene Scala, Simone Bellavia, Pier Andrea Rizzo, Jacopo Di Giovanni, Mauro Monforte, Roberta Morosetti, Giacomo Della Marca, Fabio Pilato, Aldobrando Broccolini, Paolo Profice and Giovanni Frisullo
Brain Sci. 2023, 13(11), 1541; https://doi.org/10.3390/brainsci13111541 - 2 Nov 2023
Cited by 1 | Viewed by 1635
Abstract
Background: Little evidence is available on the long-term efficacy and safety of edoxaban, mainly due to the recent release date. The primary objective of the study was to evaluate the safety of edoxaban, defined by the incidence of major bleedings. We then aimed [...] Read more.
Background: Little evidence is available on the long-term efficacy and safety of edoxaban, mainly due to the recent release date. The primary objective of the study was to evaluate the safety of edoxaban, defined by the incidence of major bleedings. We then aimed to evaluate the incidence of thromboembolic events and the persistence of edoxaban therapy in the long-term. Methods: In this observational cohort study, we included ischemic stroke patients enrolled in a previous study to evaluate the safety and efficacy of long-term edoxaban treatment. Data were collected by a trained investigator through a structured telephone interview. Results: Sixty-three subjects (median age 81.0 (73.5–88.0) years, 38.1% male) were included in the study, with a mean follow-up of 4.4 ± 0.7 years (range: 3.2–5.5 years). Only one patient (1.6%, 0.4%/year) presented a major extracranial bleeding, and none had cerebral hemorrhage. Six thromboembolic events occurred in five patients (7.9%): three recurrent strokes, two transient ischemic attacks, and one myocardial infarction (2.2%/year). Over a follow-up period of more than three years, 13 patients discontinued edoxaban (20.6%). Conclusions: Edoxaban seems to be effective and safe in the long-term. The persistence rate of edoxaban therapy is optimal after more than three years of treatment. Full article
(This article belongs to the Topic Diagnosis and Management of Acute Ischemic Stroke)
Show Figures

Figure 1

Figure 1
<p>The flow diagram of the enrollment process. Abbreviations: SATES, Prospective Observational Study of Safety of Early Treatment with Edoxaban in Patients with Ischemic Stroke and Atrial Fibrillation.</p>
Full article ">Figure 2
<p>(<b>A</b>) A graphic description of the proportion of patients who persisted and those who did non persist on edoxaban therapy at long-term follow-up, reporting, within the second group, the proportion of patients who interrupted any kind of anticoagulant therapy and those who switched to other anticoagulants; (<b>B</b>) causes of switching to other anticoagulants and their frequency; (<b>C</b>) a list of anticoagulants chosen for switching to with the relative proportions.</p>
Full article ">Figure 3
<p>(<b>A</b>) A graphic representation of the various causes of hospitalizations after the index event and their frequencies; (<b>B</b>) a figure depicting the frequencies of the causes of the death in the study sample.</p>
Full article ">
11 pages, 1873 KiB  
Article
Left Atrial Appendage Occlusion versus Novel Oral Anticoagulation for Stroke Prevention in Atrial Fibrillation—One-Year Survival
by Shmuel Tiosano, Ariel Banai, Wesam Mulla, Ido Goldenberg, Gabriella Bayshtok, Uri Amit, Nir Shlomo, Eyal Nof, Raphael Rosso, Michael Glikson, Victor Guetta, Israel Barbash and Roy Beinart
J. Clin. Med. 2023, 12(20), 6693; https://doi.org/10.3390/jcm12206693 - 23 Oct 2023
Cited by 1 | Viewed by 1244
Abstract
Aim To compare the 1-year survival rate of patients with atrial fibrillation (AF) following left atrial appendage occluder (LAAO) implantation vs. treatment with novel oral anticoagulants (NOACs). Methods: We have conducted an indirect, retrospective comparison between LAAO and NOAC registries. The LAAO registry [...] Read more.
Aim To compare the 1-year survival rate of patients with atrial fibrillation (AF) following left atrial appendage occluder (LAAO) implantation vs. treatment with novel oral anticoagulants (NOACs). Methods: We have conducted an indirect, retrospective comparison between LAAO and NOAC registries. The LAAO registry is a national prospective cohort of 419 AF patients who underwent percutaneous LAAO between January 2008 and October 2015. The NOACs registry is a multicenter prospective cohort of 3138 AF patients treated with NOACs between November 2015 and August 2018. Baseline patient characteristics were retrospectively collected from coded diagnoses of hospitalization and outpatient clinic notes. Follow-up data was sorted from coded diagnoses and the national civil registry. Subjects were matched according to propensity score. Baseline characteristics were compared using Chi-Square and student’s t-test. Survival analysis was performed using Kaplan-Meier survival curves, log-rank test, and multivariable Cox regression, adjusting for possible confounding variables. Results: This study included 114 subjects who underwent LAAO implantation and 342 subjects treated with NOACs. The mean age of participants was 77.9 ± 7.44 and 77.1 ± 11.2 years in the LAAO and NOAC groups, respectively (p = 0.4). The LAAO group had 70 (61%) men compared to 202 (59%) men in the NOAC group (p = 0.74). No significant differences were found in baseline comorbidities, renal function, or CHA2DS2-VASc score. One-year mortality was observed in 5 (4%) patients and 32 (9%) patients of the LAAO and NOAC groups, respectively. After adjusting for confounders, LAAO was significantly associated with a lower risk for 1-year mortality (HR 0.38, 95%CI 0.14–0.99). In patients with impaired renal function, this difference was even more prominent (HR 0.21 for creatinine clearance (CrCl) < 60 mL/min). Conclusions: In a pooled analysis of two registries, we found a significantly lower risk for 1-year mortality in patients with AF who were implanted with LAAO than those treated with NOACs. This finding was more prominent in patients with impaired renal function. Future prospective direct studies should further investigate the efficacy and adverse effects of both treatment strategies. Full article
(This article belongs to the Special Issue Cardiac Electrophysiology: Clinical Advances and Practice Updates)
Show Figures

Figure 1

Figure 1
<p>Kaplan-Meier curves comparing 1-year mortality of LAAO vs. NOACs. LAAO: Left Atrial Appendage Occlusion; NOACs: Novel Oral Anticoagulants.</p>
Full article ">Figure 2
<p>Kaplan-Meier curves comparing 1-year mortality of LAAO vs. NOACs by subgroups. LAAO: Left Atrial Appendage Occlusion; NOACs: Novel Oral Anticoagulant.</p>
Full article ">Figure 2 Cont.
<p>Kaplan-Meier curves comparing 1-year mortality of LAAO vs. NOACs by subgroups. LAAO: Left Atrial Appendage Occlusion; NOACs: Novel Oral Anticoagulant.</p>
Full article ">Figure 3
<p>Subgroup analysis of factors associated with 1-year mortality for those who underwent LAAO implantation. LAAO: Left Atrial Appendage Occlusion; OR: Odds Ratio; CI: Confidence Interval; y: Years; CrCl: Creatinine Clearance.</p>
Full article ">
9 pages, 752 KiB  
Article
Anticoagulation Status and Left Atrial Appendage Occlusion Indications in Hospitalized Cardiology Patients with Atrial Fibrillation: A Hellenic Cardiorenal Morbidity Snapshot (HECMOS) Sub-Study
by Dimitris Tsiachris, Panteleimon E. Papakonstantinou, Ioannis Doundoulakis, Panagiotis Tsioufis, Michail Botis, Kyriakos Dimitriadis, Ioannis Leontsinis, Athanasios Kordalis, Christos-Konstantinos Antoniou, Emmanouil Mantzouranis, Panagiotis Iliakis, Panayotis K. Vlachakis, Konstantinos A. Gatzoulis and Konstantinos Tsioufis
Medicina 2023, 59(10), 1881; https://doi.org/10.3390/medicina59101881 - 23 Oct 2023
Viewed by 1647
Abstract
Background and Objectives: The proper use of oral anticoagulants is crucial in the management of non-valvular atrial fibrillation (AF) patients. Left atrial appendage closure (LAAC) may be considered for stroke prevention in patients with AF and contraindications for long-term anticoagulant treatment. We [...] Read more.
Background and Objectives: The proper use of oral anticoagulants is crucial in the management of non-valvular atrial fibrillation (AF) patients. Left atrial appendage closure (LAAC) may be considered for stroke prevention in patients with AF and contraindications for long-term anticoagulant treatment. We aimed to assess anticoagulation status and LAAC indications in patients with AF from the HECMOS (Hellenic Cardiorenal Morbidity Snapshot) survey. Materials and Methods: The HECMOS was a nationwide snapshot survey of cardiorenal morbidity in hospitalized cardiology patients. HECMOS used an electronic platform to collect demographic and clinically relevant information from all patients hospitalized on 3 March 2022 in 55 different cardiology departments. In this substudy, we included patients with known AF without mechanical prosthetic valves or moderate-to-severe mitral valve stenosis. Patients with prior stroke, previous major bleeding, poor adherence to anticoagulants, and end-stage renal disease were considered candidates for LAAC. Results: Two hundred fifty-six patients (mean age 76.6 ± 11.7, 148 males) were included in our analysis. Most of them (n = 159; 62%) suffered from persistent AF. The mean CHA2DS2-VASc score was 4.28 ± 1.7, while the mean HAS-BLED score was 1.47 ± 0.9. Three out of three patients with a a CHA2DS2-VASc score of 0 or 1 (female) were inappropriately anticoagulated. Sixteen out of eighteen patients with a CHA2DS2-VASc score 1 or 2 (if female) received anticoagulants. Thirty-one out of two hundred thirty-five patients with a CHA2DS2-VASc score > 1 or 2 (if female) were inappropriately not anticoagulated. Relative indications for LAAC were present in 68 patients with NVAF (63 had only one risk factor and 5 had two concurrent risk factors). In detail, 36 had a prior stroke, 17 patients had a history of major bleeding, 15 patients reported poor or no adherence to the anticoagulant therapy and 5 had an eGFR value < 15 mL/min/1.73 m2 for a total of 73 risk factors. Moreover, 33 had a HAS-BLED score ≥ 3. No LAAC treatment was recorded. Conclusions: Anticoagulation status was nearly optimal in a high-thromboembolic-risk population of cardiology patients who were mainly treated using NOACs. One out of four AF patients should be screened for LAAC. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

Figure 1
<p>Anticoagulation therapy in different thromboembolic-risk AF patients according to the CHA2DS2-VASc score (AT: anticoagulation therapy).</p>
Full article ">Figure 2
<p>Potential candidates for left atrial appendage closure (LAAC). Causes for LAAC indication (third column) are non-mutually exclusive, i.e., five patients had two factors concomitantly (three experienced major bleeding plus end-stage renal disease and two experienced major bleeding plus stroke while on anticoagulant medication).</p>
Full article ">
7 pages, 1087 KiB  
Brief Report
Two Approaches to Triple Antithrombotic Therapy in Patients with Acute Coronary Syndrome and Non-Valvular Atrial Fibrillation Treated with Percutaneous Coronary Intervention: Which Is More Efficient and Safer?
by Witold Bachorski, Jakub Bychowski, Marcin Gruchała and Miłosz Jaguszewski
Diagnostics 2023, 13(19), 3055; https://doi.org/10.3390/diagnostics13193055 - 26 Sep 2023
Viewed by 982
Abstract
Introduction: Patients with acute coronary syndrome (ACS) and atrial fibrillation (AF) treated with percutaneous coronary intervention (PCI) are at high risk of bleeding and thromboembolic events. Thus, optimal treatment strategies in this challenging subset have been controversial. Herein, we aim to investigate different [...] Read more.
Introduction: Patients with acute coronary syndrome (ACS) and atrial fibrillation (AF) treated with percutaneous coronary intervention (PCI) are at high risk of bleeding and thromboembolic events. Thus, optimal treatment strategies in this challenging subset have been controversial. Herein, we aim to investigate different triple antithrombotic treatment (TAT) strategies in patients with ACS and AF after PCI. Methods: This was a retrospective, single-center study based on all consecutive patients with the diagnosis of ACS and AF treated with vitamin K antagonists (VKA) or non-vitamin K antagonist oral anticoagulants (NOAC) plus dual antiplatelet therapy using a P2Y12 inhibitor (clopidogrel) and aspirin (for 1 to 3 months) and observed for 12 months for major adverse cardiac events (MACE) and major or clinically relevant non-major bleeding incidents. Results: MACE occurred in 26.6% of patients treated with the VKA and 30.9% with NOAC (p = 0.659). Bleeding occurred in 7.8% of patients treated with VKA and 7.4% with NOAC (ns). Conclusions: Among patients with ACS and AF who had undergone PCI, there was no significant difference in the risk of bleeding and ischemic events among those who received TAT with NOAC and VKA. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Flow chart for study. ACS—acute coronary syndrome, AF—atrial fibrillation, SAT—single antithrombotic therapy, DAT—double antithrombotic therapy, and TAT—triple antithrombotic therapy.</p>
Full article ">Figure 2
<p>Kaplan–Meier survival curve for major adverse cardiac events (MACE) (<b>A</b>). Kaplan–Meier survival curve for bleeding events (<b>B</b>).</p>
Full article ">
12 pages, 1395 KiB  
Systematic Review
Outcomes and Safety of Direct Oral Anticoagulants (DOACs) versus Vitamin K Antagonists (VKAs) amongst Patients with Valvular Heart Disease (VHD): A Systematic Review and Meta-Analysis
by Ghanshyam Patel, Beshoy Iskandar, Nikhila Chelikam, Siddhant Jain, Vandit Vyas, Tanvi Singla, Lavanya Dondapati, Ali Bombaywala, Appala Suman Peela, Milan Khealani, Sindhu Mukesh, Hariprasad Reddy Korsapati, Aishwarya Reddy Korsapati, Henok Regassa, Nitesh Jain, Urvish Patel and Vikramaditya Samala Venkata
Hearts 2023, 4(3), 61-72; https://doi.org/10.3390/hearts4030008 - 7 Sep 2023
Viewed by 1650
Abstract
Background: Both valvular heart disease (VHD) and atrial fibrillation (AF) frequently coexist. AF is an important cause of arrhythmias with a definitive cardiovascular morbidity. The use of either vitamin K antagonists (VKAs/warfarin) or direct oral anticoagulants (DOACs) (also known as new oral anticoagulants [...] Read more.
Background: Both valvular heart disease (VHD) and atrial fibrillation (AF) frequently coexist. AF is an important cause of arrhythmias with a definitive cardiovascular morbidity. The use of either vitamin K antagonists (VKAs/warfarin) or direct oral anticoagulants (DOACs) (also known as new oral anticoagulants (NOACs)) has been the mainstay for preventing stroke and systemic embolism in patients with VHD and/or AF, and this has been broadly discussed. However, there are limited studies on anticoagulation therapy for patients with valvular atrial fibrillation (VAF). The main aim of this meta-analysis was to evaluate the outcomes (stroke–vascular events and intracranial bleeding) following DOAC and VKA treatment amongst patients with VAF. Methods: We identified clinical trials and observational studies published in the last 10 years. A systematic review and a meta-analysis were performed to evaluate the outcomes of patients with valvular atrial fibrillation following DOAC vs. VKA treatment. Data evaluation was performed using Review Manager 5.4; the endpoints were stroke–vascular events and intracranial bleeding following DOAC and VKA treatment amongst VAF patients. Risk ratios (RR) were evaluated with 95% confidence intervals. Using random effects models, forest plots were obtained. Heterogeneity was assessed by using the I2 statistic. Results: Eight studies were included in this metanalysis, and a total of fifteen thousand two hundred and fifteen patients (DOAC (8732) and VKA (6483)) were pooled. We found a significant risk reduction in stroke–vascular events when using DOACs in comparison with using VKAs (pooled RR: 0.76; 95% CI: 0.64–0.90, p = 0.002). A total of 14862 patients (DOAC (8561) and VKA (6301)) were pooled from a total of six studies for intracranial bleeding. We found a significant risk reduction in terms of intracranial bleeding when using DOACs in comparison with using VKAs (pooled RR: 0.43; 95% CI: 0.24–0.77, p ≤ 0.05). Conclusions: When compared to VKAs, DOAC agents were found to have less risk of stroke–vascular events and intracranial bleeding. Further prospective studies are essential to establish the efficacy and safety of DOAC agents in patients with various subtypes of VAF. Full article
Show Figures

Figure 1

Figure 1
<p>Study Selection Process.</p>
Full article ">Figure 2
<p>Forest plot for risk of stroke–vascular events after DOAC therapy.</p>
Full article ">Figure 3
<p>Forest plot for all-cause mortality after DOAC therapy.</p>
Full article ">Figure 4
<p>Forest plot for major bleeding after DOAC therapy.</p>
Full article ">Figure 5
<p>Forest plot for intracranial bleeding after DOAC therapy.</p>
Full article ">Figure 6
<p>Forest plot for composite poor outcome after DOAC therapy.</p>
Full article ">
16 pages, 1084 KiB  
Article
Plasma Drug Values of DOACs in Patients Presenting with Gastrointestinal Bleeding: A Prospective Observational Study
by Dorotea Bozic, Damir Alicic, Dinko Martinovic, Ivan Zaja, Josipa Bilandzic-Ivisic, Rosana Sodan, Branka Kresic, Andre Bratanic, Zeljko Puljiz, Zarko Ardalic and Josko Bozic
Medicina 2023, 59(8), 1466; https://doi.org/10.3390/medicina59081466 - 16 Aug 2023
Viewed by 1243
Abstract
Background and Objectives: Anticoagulants are a well-known risk factor for gastrointestinal bleeding (GIB). In recent years, direct oral anticoagulants (DOACs) have taken a leading role in the treatment and prevention of thromboembolic incidents. The aim of this study was to investigate the [...] Read more.
Background and Objectives: Anticoagulants are a well-known risk factor for gastrointestinal bleeding (GIB). In recent years, direct oral anticoagulants (DOACs) have taken a leading role in the treatment and prevention of thromboembolic incidents. The aim of this study was to investigate the prevalence of DOAC-treated patients with GIB whose plasma drug concentrations exceeded the cut-off values reported in the literature and to evaluate their clinical characteristics. Materials and Methods: Patients who were admitted to the Intensive Care Unit in the period 2/2020–3/2022 due to GIB were prospectively included in the study and classified into three groups according to the prescribed type of DOAC (apixaban, rivaroxaban, and dabigatran). For all participants, it was determined if the measured plasma drug levels exceeded the maximum serum concentration (Cmax) or trough serum concentration (Ctrough) obtained from the available data. A comparison of clinical parameters between the patients with and without excess drug values was performed. Results: There were 90 patients (54.4% men) included in the study, of whom 27 were treated with dabigatran, 24 with apixaban, and 39 with rivaroxaban. According to Cmax, there were 34 (37.8%), and according to Ctrough, there were 28 (31.1%) patients with excess plasma drug values. A statistically significant difference regarding excess plasma drug values was demonstrated between DOACs according to both Cmax (p = 0.048) and Ctrough (p < 0.001), with the highest rate in the group treated with dabigatran (55.6% for Cmax and 59.3% for Ctrough). Multivariate logistic regression showed that age (OR 1.177, p = 0.049) is a significant positive and glomerular filtration rate (OR 0.909, p = 0.016) is a negative predictive factor for excess plasma drug values. A total of six (6.7%) patients had fatal outcomes. Conclusions: Plasma drug concentrations exceed cut-off values reported in the literature in more than one-third of patients with GIB taking DOAC, with the highest rate in the dabigatran group. Clinicians should be more judicious when prescribing dabigatran to the elderly and patients with renal failure. In these patients, dose adjustment, plasma drug monitoring, or substitution with other, more appropriate DOACs should be considered. Full article
(This article belongs to the Section Cardiology)
Show Figures

Scheme 1

Scheme 1
<p>Distribution of patients in the study sample (N = 90) regarding their (<b>A</b>) C<sub>max</sub> and (<b>B</b>) C<sub>trough</sub> measurements.</p>
Full article ">Scheme 2
<p>Comparison of the frequency (%) of patients with excess plasma drug concentrations between dabigatran (N = 27), apixaban (N = 24) and rivaroxaban (N = 39) according to the (<b>A</b>) C<sub>max</sub> and (<b>B</b>) C<sub>trough</sub> measurements. * chi-square test.</p>
Full article ">Scheme 2 Cont.
<p>Comparison of the frequency (%) of patients with excess plasma drug concentrations between dabigatran (N = 27), apixaban (N = 24) and rivaroxaban (N = 39) according to the (<b>A</b>) C<sub>max</sub> and (<b>B</b>) C<sub>trough</sub> measurements. * chi-square test.</p>
Full article ">
15 pages, 2651 KiB  
Article
Improved Process for the Synthesis of 3-(3-Trifluoromethylphenyl)propanal for More Sustainable Production of Cinacalcet HCl
by Vikas Damu Rathod, Stefano Paganelli, Marijan Kočevar, Marko Krivec and Oreste Piccolo
Molecules 2023, 28(16), 6042; https://doi.org/10.3390/molecules28166042 - 13 Aug 2023
Viewed by 1291
Abstract
Cinacalcet (I), sold as hydrochloride salt, is a calcimimetic drug which has been approved for the treatment of secondary hyperparathyroidism in patients with chronic renal disease and for the treatment of hypercalcemia in patients with parathyroid carcinoma. Here, an improved method [...] Read more.
Cinacalcet (I), sold as hydrochloride salt, is a calcimimetic drug which has been approved for the treatment of secondary hyperparathyroidism in patients with chronic renal disease and for the treatment of hypercalcemia in patients with parathyroid carcinoma. Here, an improved method for the synthesis of 3-(3-trifluoromethylphenyl)propanal (II), a key intermediate for the preparation of I, is described. The protocol required a Mizoroki–Heck cross-coupling reaction between 1-bromo-3-(trifluoromethyl)benzene and acroleine diethyl acetal, catalyzed by Pd(OAc)2 in the presence of nBu4NOAc (tetrabutylammonium acetate), followed by the hydrogenation reaction of the crude mixture of products in a cascade process. Palladium species, at the end of the reaction, were efficiently recovered as Pd/Al2O3. The procedure was developed under conventional heating conditions as well as under microwave-assisted conditions. The obtained mixture of 1-(3,3-diethoxypropyl)-3-(trifluoromethyl)benzene (III), impure for ethyl 3-(3-trifluoromethylphenyl) propanoate (IV), was finally treated, under mild conditions, with potassium diisobutyl-tert-butoxyaluminum hydride (PDBBA) to obtain after hydrolysis 3-(3-trifluoromethylphenyl)propanal (II), in an excellent overall yield and very high purity. Microwave conditions permitted a reduction in reaction times without affecting selectivity and yield. The final API was obtained through reductive amination of (II) with (R)-(+)-1-(1-naphthyl)ethylamine (V) using a catalyst prepared by us with a very low content of precious metal. Full article
(This article belongs to the Section Organic Chemistry)
Show Figures

Figure 1

Figure 1
<p>Structure of Cinacalcet (<b>I</b>).</p>
Full article ">Scheme 1
<p>Reaction scheme for the synthesis of Cinacalcet (<b>I</b>).</p>
Full article ">Scheme 2
<p>Mizoroki–Heck cross-coupling reaction.</p>
Full article ">Scheme 3
<p>Hypothesized reaction mechanism.</p>
Full article ">Scheme 4
<p>Hydrogenation of the mixture <b>VI</b> + <b>IV</b> and hydrolysis to afford the mixture <b>II</b> + <b>IV</b>.</p>
Full article ">Scheme 5
<p>Purification of <b>II</b> via Bertagnini adduct.</p>
Full article ">Scheme 6
<p>Reduction of <b>IV</b> to <b>II</b>.</p>
Full article ">Scheme 7
<p>Reductive amination of <b>II</b>.</p>
Full article ">
14 pages, 1788 KiB  
Article
Anticoagulant Treatment in Patients with AF and Very High Thromboembolic Risk in the Era before and after the Introduction of NOAC: Observation at a Polish Reference Centre
by Bernadetta Bielecka, Iwona Gorczyca-Głowacka, Agnieszka Ciba-Stemplewska and Beata Wożakowska-Kapłon
Int. J. Environ. Res. Public Health 2023, 20(12), 6145; https://doi.org/10.3390/ijerph20126145 - 16 Jun 2023
Viewed by 1470
Abstract
Atrial fibrillation (AF) is associated with an increased risk of stroke. Therefore, patients with AF require appropriate management and anticoagulant therapy. To balance therapy risks and benefits, oral anticoagulants (OAC) treatment should be ‘tailored’ in patients at a high risk of stroke and [...] Read more.
Atrial fibrillation (AF) is associated with an increased risk of stroke. Therefore, patients with AF require appropriate management and anticoagulant therapy. To balance therapy risks and benefits, oral anticoagulants (OAC) treatment should be ‘tailored’ in patients at a high risk of stroke and bleeding. However, some studies have demonstrated that certain groups of patients do not receive anticoagulants despite the high risk of stroke or thromboembolism. The study aimed to analyse therapeutic methods of stroke prevention in very high-risk patients (CHA2DS2-VASc score of ≥5 in men and ≥6 in women), identify factors predisposing against the use of OACs and assess the administration of anticoagulants before the introduction of non-vitamin K antagonist OAC (NOAC) in 2004–2011 and beyond (years 2012–2019). The analysis covered 2441 patients with AF at a very high thromboembolic risk who were hospitalised in a reference cardiological centre from 2004 to 2019. Data concerning patients’ sex, age, comorbidities, type of AF, renal and echocardiographic parameters, reasons for hospitalisation and applied treatment were collected from medical records. HAS-BLED, CHADS2, and CHA2DS2-VASc scores were calculated for all patients. The treatment with oral anticoagulants was compared in the entire population over 2004–2011 and 2012–2019. In this study, a fifth of patients were not treated with OAC. Most patients hospitalised in the years 2012–2019 were treated with OAC. The predictors of not using OAC turned out to be: age of >74 years, heart failure, cancer, paroxysmal AF, and acute coronary syndrome (ACS) or elective coronary angiography/percutaneous coronary intervention (PCI) as a reason for hospitalisation. The introduction of NOAC was associated with a decline in the use of VKA (from 62% to 19.1%) and APT (from 29.1% to 1.3%). This study outlines reasons to initiate OAC treatment in very high-risk patients in clinical practice. Full article
Show Figures

Figure 1

Figure 1
<p>The flow chart of the study. Abbreviations: AF, atrial fibrillation; APT, antiplatelet drugs; OAC, oral anticoagulant therapy; LMWH, low molecular weight heparin.</p>
Full article ">Figure 2
<p>The prescription of OAC based on the CHA2DS2-VASc score in the entire population. Abbreviation: OAC, oral anticoagulant.</p>
Full article ">Figure 3
<p>The prescription of OAC based on the HAS-BLED score in the entire population. Abbreviation: OAC, oral anticoagulant.</p>
Full article ">Figure 4
<p>Stroke prophylaxis in patients with a very high risk of thromboembolic events with AF. Abbreviation: APT, antiplatelet drug; LMWH, low mass weight heparin; NOAC, non-vitamin K antagonist oral anticoagulants; VKA, vitamin K antagonists.</p>
Full article ">
Back to TopTop