Next Article in Journal
First Report of a Low-Frequency Mosaic Mutation in the Hydroxymethylbilane Synthase Gene Causing Acute Intermittent Porphyria
Previous Article in Journal
Study of Optimal Conditions to Grow Thai Ganoderma, Fruiting Test, Proximate and Their Alpha Glucosidase Inhibitory Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Inflammatory and Anticancer Effects of Anticoagulant Therapy in Patients with Malignancy

1
Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
2
Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
3
Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, NA, Italy
4
Cardiology Unit, Umberto I Hospital, 84014 Nocera Inferiore, SA, Italy
*
Author to whom correspondence should be addressed.
Life 2023, 13(9), 1888; https://doi.org/10.3390/life13091888
Submission received: 14 August 2023 / Revised: 7 September 2023 / Accepted: 8 September 2023 / Published: 10 September 2023
(This article belongs to the Section Physiology and Pathology)

Abstract

:
Optimizing the anticoagulation therapy is of pivotal importance in patients with a malignant tumor, as venous thromboembolism (VTE) has become the second-leading cause of death in this population. Cancer can highly increase the risk of thrombosis and bleeding. Consequently, the management of cancer-associated VTE is complex. In recent years, translational research has intensified, and several studies have highlighted the role of inflammatory cytokines in cancer growth and progression. Simultaneously, the pleiotropic effects of anticoagulants currently recommended for VTE have emerged. In this review, we describe the anti-inflammatory and anticancer effects of both direct oral anticoagulants (DOACs) and low-molecular-weight heparins (LWMHs).

1. Introduction

Cancer patients are at a high risk for venous thromboembolism (VTE). After being diagnosed with a malignant tumor, the thrombotic risk increases quickly and remains high over time [1]. The advances in cancer treatment and earlier diagnosis have contributed to a longer life expectancy, and cancer-related thromboembolic events have emerged as the second-leading cause of death [2]. Peculiar features of VTE in cancer patients can possibly explain the data concerning the morbidity and mortality. The incidence of bleeding and recurrent VTE, despite appropriate anticoagulation, is high. Additionally, VTE events are linked to more hospitalizations and can hinder the delivery of lifesaving treatments.
Traditionally, vitamin K antagonists (VKAs) were used in conjunction with rapid-acting parenteral anticoagulants such as low-molecular-weight heparins (LWMHs), fondaparinux, or unfractionated heparin (UFH) to treat acute VTE. However, this strategy presented challenges in cancer patients due to many drug–drug interactions (DDIs) that either attenuated or amplified the anticoagulant effect of VKAs, as well as variability in gastrointestinal (GI) absorption caused by vomiting, diarrhea, or malnutrition. VKAs have a long half-life, which can make therapy more challenging in the context of thrombocytopenia or surgery, common conditions among cancer patients. In the early 2000s, for the first time, a randomized controlled trial (RCT) showed the superiority of dalteparin monotherapy versus warfarin in preventing recurrent VTE in cancer patients, without increasing the bleeding risk [3]. Recently, a meta-analysis, including twenty-three RCTs with 6980 patients, established the beneficial effects of LWMH over VKAs [4]. Nevertheless, LWMHs have some limitations such as high cost, uncomfortable route of administration, and reduced adherence to a long-term treatment [5].
Direct oral anticoagulants (DOACs) have overcome the drawbacks of both VKAs and parenteral LWMHs, i.e., oral administration without the need for routine blood monitoring, good therapeutic adherence, and fewer intracranial hemorrhages. Based on this evidence, DOACs are considered the first choice when anticoagulation therapy is needed in different clinical scenarios [6,7,8,9], even in that subgroups of patients under-represented in the RCTs [10,11,12,13,14,15,16,17,18,19,20,21,22,23]. Recently, DOACs were tested in cancer patients [24,25,26,27,28], and according to the international guidelines, rivaroxaban, apixaban, and edoxaban are considered the first-line anticoagulant therapy for patients with malignancy, except for those with GI and genitourinary cancers [29,30]. Previous experimental studies suggested “pleiotropic” effects on inflammation pathway and cancer-progression of both parenteral and oral anticoagulant therapies. The present narrative review aimed to describe anti-inflammatory and anticancer effects of anticoagulant therapy in patients with malignancy.

2. Cancer, Inflammation, and Thrombosis

2.1. Role of Inflammation in Cancer

Inflammation is a protective process in vascularized tissues that acts against foreign bodies, pathogens, or injuries. It serves to rid the host of both the initial cause of cell injury, i.e., microbes or toxins, and necrotic byproducts of inflammatory response. The mediators of inflammation include phagocytic leukocytes, antibodies, complement proteins, chemokines, and cytokines. Most of these normally circulate in an inactive state in the blood, from which they can be rapidly recruited to the damaged tissue. Inflammation typically develops through five sequential steps: (1) recognition of the noxious agent by receptors on sentinel cells, such as tissue macrophages or mast cells, resulting in the activation of these cells and subsequent release of inflammatory mediators, which trigger an immune response; (2) recruitment of leukocytes and plasma proteins into the tissues; (3) removal of pathogenic noxa mainly by phagocytic cells, which ingest and destroy microbes and dead cells; (4) regulation of the response involves resolution of inflammation once it has achieved its purpose; and (5) repair of damaged tissues. Cytokines are the main signaling molecules released by inflammatory cells and work at different steps in the inflammation pathway. They are classified into proinflammatory cytokines, such as Interleukin-1 (IL-1), IL-6, IL-15, IL-17, IL-23, tumor necrosis factor alpha (TNF-α), and interferon gamma (IFN-γ), and anti-inflammatory cytokines, such as IL-4, IL-10, IL-13, and transforming growth factor beta (TGF-β). C-C motif chemokine ligand 2 (CCL2) and C-X-C motif chemokine 12 (CXCL12) are small molecules necessary for the recruitment of inflammatory cells to the inflammatory area [31]. Rudolf Virchow was the first to describe a correlation between cancer and inflammation in 1863, noting that tumors arose at sites of chronic inflammation and that inflammation caused increased cell proliferation [32]. On one hand, inflammation has a protective role against cancer by mediating the direct elimination of mutated cells and enhancing the response to anticancer therapies. On the other hand, chronic, dysregulated, persistent, and unresolved inflammation has been associated with an increased risk of malignancy, as well as the malignant progression of cancer in most types of cancer. In the inflammatory tumor microenvironment (TME), there is an increase in DNA and protein damage, activation of oncogenes, and release of ROS which ultimately affect multiple signaling pathways such as nuclear factor-kB (NF-kB), Kirsten rat sarcoma virus (K-RAS), Janus kinase/signal transducer, and activator of transcription 3 (JAK/STAT3). These activated transcription factors mediate the expression of key cytokines and chemokines, including TNF-α and IL-6, as well as other proinflammatory enzymes. Cyclooxygenase 2 (COX-2) is upregulated in several malignancies. COX-2-generated prostaglandins enhance the migration of cancer cells while limiting apoptosis. Additionally, they promote neo-angiogenesis. In TME, cancer cells can not only prevent dendritic cells from presenting tumor-associated antigens but also recruit many immunosuppressive cells. In turn, these immunosuppressive cells provide a rich proangiogenic and pro-tumoral microenvironment, preventing both innate and specific immune response. Autocrine and paracrine effects of the cytokines released within the tumor microenvironment might further sustain these cells [33,34]. Several studies have shown that up to 25% of cancers are related to chronic inflammatory diseases [35]. Plenty of chronic inflammatory diseases are considered fully fledged precancerous environments. Therefore, targeting inflammation has become extremely important. However, a thorough understanding of the underlying molecular pathways is crucial for the development of targeted therapies.
Cytokines and chemokines have a direct effect on cancer cells and contribute to the stimulation of epithelial-to-mesenchymal transition (EMT), promoting metastases. TNF-α stimulates cell growth, survival, invasion, metastasis, and neo-angiogenesis. IL-6 has both local and systemic tumorigenic actions mediated by JAK/STAT3 pathway. Janus kinases are a family of four intracellular nonreceptor tyrosine kinases (JAK1, JAK2, JAK3, and TYK2) that act as primary signal transducers from cell surface receptors activated by cytokines and growth factors. These kinases in turn phosphorylate STAT proteins on tyrosine residues, enhancing their function as transcription factors in the expression of several genes involved in cell survival.
IL-6 seems to accelerate cell proliferation by activating inflammasome via JAK2/STAT3/SRY-Box Transcription Factor 4 (Sox4)/NOD-like receptor protein 3 (NLRP3) pathways both in vitro and in vivo [36].
IL-1α is associated with a less differentiated and more aggressive cancer through the activation of vascular endothelium and infiltration by tumorigenic inflammatory cells. IL-8 promotes tumorigenesis by inducing migration of tumor cells, promoting angiogenesis, and enhancing metastasis in patients with melanoma. Chemokine receptors are highly expressed by several malignancies and facilitate the spread of cancer cells throughout the body [37].
Several studies have found that monocyte chemotactic protein-1 (MCP-1) expression in TME is associated with tumor development, tumor invasion and metastasis, angiogenesis, and immune cell infiltration. MCP-1 exerts its effects mainly via the MCP-1/CC2 axis and leads to the activation of classical signaling pathways, such as Phosphoinositide 3-kinase (PI3K)/Protein kinase B (PKB, or Akt)/Mammalian Target of Rapamycin (mTOR), Extracellular signal-regulated kinase (ERK)/Glycogen synthase kinase 3 beta (GSK-3β)/Snail, RAF proto-oncogene serine/threonine-protein kinase (c-Raf)/Mitogen-activated protein kinase kinase (MEK)/ERK, and Mitogen-activated protein kinases (MAPKs) in different cells [38]. The NF-κB transcription factor family has a major role in inflammation. It is activated by canonical and non-canonical signaling pathways, which differ in both signaling components and biological functions [39]. Activation of non-canonical pathways has been shown to support tumor progression in multiple cancers, both solid and hematological [40]. In most tumors, NF-κB is constitutively active and activates antiapoptotic and cell-cycle progression genes, such as B cell leukemia/lymphoma 2 (BCL2), and promotes the expression of the hypoxia-inducible factor-1α (HIF-1α) and favors cell migration and invasion [34].

2.2. Role of Inflammation in Thrombosis

Thrombosis, inflammation, and cancer are disjointed physiological processes with a deep interdependence [41]. Circulating blood platelets are crucial to each process. Recent studies suggest that thrombosis has an important role in immune defense. Hence, the concept of immunothrombosis arose [42]. The host defense system is not limited to a strong immune response but also includes thrombosis promotion. Immunothrombosis indicates an innate immune response induced by the formation of thrombi within micro-vessels. The interaction between platelets and immune cells is supported by coagulation factors and complement system and generates an intravascular network that facilitates the recognition, containment, and destruction of pathogens [43]. When a vascular injury occurs, platelets are recruited to the site of injury by a process involving the interaction of specific platelet receptors with sub-endothelial matrix proteins [44].
After this initial phase, platelets become activated and adhere tightly to the vessel wall and release intracellular granules, which promote their own activation. The continuous processes of adhesion, activation and aggregation of platelets causes rapid growth and stabilization of thrombus [45].
Simultaneously with platelet activation, the coagulation cascade is activated at the site of the damage. The active contribution of innate immune cells to clot formation is also very important.
Platelets and clotting factors activate innate immune cells. Within a developing clot, platelets and coagulation byproducts meticulously regulate the effective functions of innate immune cells.
Indeed, monocytes and neutrophils are rapidly recruited to the endothelium and are included in growing intravascular clots. Recruitment of innate immune cells proceeds throughout the thrombus development. Platelets produce a myriad of proinflammatory cytokines and chemokines. Activated platelets also express IL-1β, which induces tissue factor (TF) expression in endothelial cells and stimulates the expression of endothelial–leukocyte adhesion molecules [46]. Additionally, IL-1β further promotes platelet activation via the IL-1 receptor. Platelets adhere to von Willebrand factor (VWF) and attach to endothelial cells, and this interaction causes tethering and rolling of leukocytes onto the endothelial surface. In this way, platelets not only lead leukocytes to a site where inflammation potentially causes atherosclerosis but also contain depots of proinflammatory mediators, such as thromboxanes and CD40 ligand [47]. An equally important role is played by proinflammatory cytokines. IL-6 and IL-1 can activate the coagulation cascade and increase fibrin deposition, through TF, and interact with antithrombin III (AT III) and protein C with a downregulating effect [48]. Moreover, coagulation proteases TF, FXa, and FIIa are key modulators of the systemic inflammatory response [49], through the activation of PARs. PARs belong to the superfamily of G-protein-coupled receptors and are located on platelets, leukocytes, and endothelial cells [50]. Four isoforms of PAR (PAR1–PAR4) have been isolated. FIIa activates PAR-1, PAR-3, and PAR-4. FXa interacts with PAR-1 and PAR-2 [51,52,53]. FXa is a vitamin-K-dependent serine protease that leads to the generation of thrombin (FIIa) from prothrombin (FII) and is a common point between the intrinsic and extrinsic coagulation pathways. Recent studies have demonstrated that FXa is also a key modulator of systemic inflammatory response, through PARs activation and NF-kB transcription [54,55]. PARs activation induces the degradation of I-κB, an inhibitor protein which causes translocation of NF-kB to the nucleus, to activate promoter regions of many proinflammatory genes. FXa increases the production of IL-6, IL-8, and some chemokines, such as MCP-1 [56,57]. The expression of adhesion molecules, such as E-selectin, intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) is upregulated, promoting leukocyte migration and recruitment during inflammation [58]. Finally, FXa fosters the pro-fibrotic response, increasing the release of TGF β, fibronectin, and platelet-derived growth factor (PDGF) by fibroblasts [59,60]. FIIa is a serine protease that promotes the conversion of fibrinogen to fibrin monomers, which, upon polymerization, form a fibrin clot. Some studies have revealed FIIa-mediated proinflammatory activities in vitro, such as the production of IL-6 and IL-8 by monocytes and of chemokines, i.e., platelet activating factor (PAF) and MCP-1, by platelets and monocytes [57,61].

3. Indications for Anticoagulant Therapy in Cancer Patients

Cancer is associated with an increased risk of both hemorrhagic and thromboembolic complications. Several factors, including patient characteristics, cancer type and location, comorbidities, adverse effects (AEs) of antineoplastic agents and drug–drug interactions (DDIs) between antineoplastic drugs and anticoagulants may influence both hemorrhagic and thromboembolic risks. Therefore, the initiation and selection of an anticoagulant regimen require careful consideration. Over the years, there have been many concerns about the use of anticoagulants in cancer patients. The recent international guidelines on cardio-oncology provide a clear path forward for different clinical conditions [62].

3.1. Treatment and Secondary Prevention of Venous Thromboembolism

Apixaban, edoxaban, or rivaroxaban are recommended for the treatment of incident or symptomatic VTE in cancer patients without contraindications such as unoperated gastrointestinal or genitourinary cancer, history of recent bleeding or within 7 days of major surgery, significant thrombocytopenia (platelet count < 50,000/µL), severe renal dysfunction (CrCl, 15 mL/min), or gastrointestinal complications (Class I; Level of Recommendation: A). LMWHs are recommended for the treatment of symptomatic VTE in cancer patients with platelet count > 50,000/µL (Class I; Level of Recommendation: A) [62]. Several RCTs and meta-analyses showed that VKAs are associated with a higher risk of recurrent VTE and similar risk of major bleeding compared to LMWHs [62]. The minimum anticoagulation regimen is six months, and it can be extended in the presence of active malignancy, metastatic disease, or chemotherapy use. However, due to high risk of bleeding, cancer patients should be subjected to a periodic assessment of the risk/benefit ratio. The duration of anticoagulant regimen in patients with catheter-associated thrombosis depends on whether the catheter is removed or remains in situ. If the catheter is removed, the anticoagulation regimen should continue for a minimum of three months and until the resolution of the thrombus is confirmed with cardiac imaging. If the catheter remains in situ, long-term therapeutic anticoagulation should be continued (Class I; Level of Recommendation: C) [62].

3.2. Primary Prevention of Venous Thromboembolism

Low-dose anticoagulation is required in patients undergoing surgery and those hospitalized or in prolonged bed rest (Class I; Level of Recommendation: B). LMWHs used as primary thromboprophylaxis for 4 weeks after a major abdominal or pelvic cancer surgery have favorable outcomes as suggested by the ENOXACAN (enoxaparin and cancer) II study (Class I; Level of Recommendation: B) [63]. No data suggest the superiority of any LMWH over the others. Ambulatory cancer patients at high risk of thrombosis receiving systemic therapy without significant contraindications may consider primary thromboprophylaxis with a direct oral anticoagulant (DOAC) (apixaban or rivaroxaban) or LMWH (Class IIb; Level of Recommendation: B). VTE risk should be individually determined using assessments such as the Khorana [64] or the COMPASS-CAT [65] score. Apixaban (2.5 mg twice a day) treatment resulted in a significantly lower rate of VTE, although the occurrence of major bleeding episodes was higher than with a placebo [66]. Rivaroxaban (10 mg once a day) therapy resulted in a non-significant lower incidence of VTE or death due to VTE with no significant differences compared to the placebo regarding bleeding risk [67].

3.3. Atrial Fibrillation

Cancer patients have a higher risk of developing atrial fibrillation (AF) [62]. In the general population, the choice to start anticoagulant therapy is based on risk stratification for stroke/systemic embolism through CHA2DS2-VASc score [68]. The CHA2DS2-VASc score has not been validated in cancer patients [69]. The CHA2DS2-VASc score in cancer patients should not be used to indicate the initiation of anticoagulant therapy based on the risk stratification for stroke/systemic embolism but to identify patients at low thrombotic risk who can avoid anticoagulation [70]. For bleeding risk assessment, the HAS-BLED score may be considered [62]. A proposed approach to anticoagulant therapy in cancer is based on the acronym T (thrombotic risk), B (bleeding risk), I (drug interactions), P (access and patient preferences) [71].
VKA use in cancer patients is limited by their drawbacks and remains indicated only in patients with a mechanical prosthetic valve or moderate to severe mitral stenosis (Class IIa; Level of Recommendation: B).
LMWHs represent a viable short-term anticoagulation option, in particular in hospitalized patients with a recent cancer diagnosis, advanced-stage cancer, or during cancer therapy [62]. However, no data suggest LMWHs’ efficacy for stroke or systemic embolism prevention in AF, and their use is only based on their proven efficacy and safety in VTE.
The use of a DOACs in cancer patients with AF has not been evaluated in a dedicated RCT. However, secondary analyses of phase III DOAC trials using direct factor Xa inhibitors [72,73,74] suggest that DOACs have better safety profiles and are not inferior to VKA on comparison [62]. DOACs should be considered for stroke prevention (excluding patients with moderate-to-severe mitral stenosis or mechanical heart valves) in patients without a high bleeding risk, significant DDIs, or severe renal disease (Class IIa; Level of Recommendation: B). Left atrial appendage (LAA) occluder devices are used in selected cancer patients (Class IIb; Level of Recommendation: C) [62].
Long-term anticoagulant therapy is recommended in adult patients with CHA2DS2-VASc score ≥2 in men or ≥3 in women and must be considered also when the score is 1 in men and 2 in women (Class I; Level of Recommendation: C) [62].

4. Anti-Inflammatory Effects of DOACs

4.1. Rivaroxaban

The potential anti-inflammatory properties of DOACs have so far been assessed mainly with rivaroxaban (Table 1) [75,76,77,78,79,80,81,82,83,84,85].
Four cellular models have been used to provide insights into the pleiotropic actions of rivaroxaban [75,76,77,78]. Proinflammatory cytokines, chemokines, and adhesion molecules were negatively modulated after rivaroxaban exposition. In human atrial cells, rivaroxaban decreased expression of PARs, IL-8, and ICAM-1 [75]. Besides a reduced PARs expression, rivaroxaban also inhibited PAR-signaling in HUVECs, abolishing transcription of chemokines, MCP-1, and tissue factor (TF) [78]. The reduced production of chemiotactic factors confirmed previous findings from Ishibashi et al. [76,77].
Five in vivo studies were conducted in different mouse models [79,80,81,82,83]. The anti-inflammatory effects were consistent across the experiments. Vascular inflammation was reduced regardless of the etiology [79,83]. Indeed, rivaroxaban prevented recruitment of inflammatory cells limiting MCP-1 and IL-6 generation. [79,83] Moreover, key inflammatory molecules (TNF-a and IL-1) or upstream transcription factors such as NF-Kb were successfully tackled in the remaining investigations [80,81,82]. Finally, two observational studies [84,86] and a post-hoc analysis of the X-VeRT RCT [85] showed that rivaroxaban reduced the inflammatory biomarkers among patients with both AF [84,86] and coronary and periphery artery disease [86].
Table 1. Anti-inflammatory effects of rivaroxaban.
Table 1. Anti-inflammatory effects of rivaroxaban.
AuthorProtocolTargetDisease ModelResults
Bukowska [75]Pre-clinical in vitroHuman atrial tissue cellsAtrial tissue cultivated with Fxa and stimulated with 4 Hz pacing↓ PARs
↓ ICAM-1
↓ IL-8
Ishibashi [76]Pre-clinical in vitroHUVECsCitrated human plasma-induced ROS generation and adhesion molecules expression↓ MCP-1
↓ ICAM-1
Ishibashi [77]Pre-clinical in vitroHuman proximal tubular cellsAGE exposition↓ MCP-1
Ellinghaus [78]Pre-clinical in vitroHUVECsThrombin exposition and recalcified plasma incubation↓ VCAM-1, ICAM-1, MCP-1
↓ IL-8
↓ CXCL1, CXCL2
↓ TF
Zhou [79]Pre-clinical in vivoApoE-deficient miceAtherosclerosis of innominate artery↓ MCP-1
↓ IL-6, TNF-α
Hara [80]Pre-clinical in vivoC57BL/6 miceAtherosclerosis of aortic arch↓ IL-1β, TNF-α
Imano [81]Pre-clinical in vivoMale C57BL/6 J miceIntermittent hypoxia↓ NF-kB
Daci [82]Pre-clinical in vivoWistar ratsLPS-induced acute inflammation↓ MCP-1
↓ IL-6
Abdelzaher [83]Pre-clinical in vivoAdult male Wistar ratsCUMS-induced depression↓ NF-kB
Martins [84]Observational study127 AF patients-↓ IL-2, IL-4, IL-10, TNF-α
Kirchhof [85]Post-hoc analysis of RCT918 AF patients-↓ IL-6
Russo [87]Observational study44 CAD/PAD patients-↓ IL-6 ↓ fibrinogen
PARs: protease-activated receptors; ICAM-1: intercellular adhesion molecule-1; IL-8: Interleukin-8; HUVECs: human umbilical vein endothelial cells; MCP-1: monocyte chemoattractant protein-1; VCAM-1: vascular adhesion molecule-1, CXCL: C-X-C motif chemokine ligand; TF: tissue factor; ApoE: Apolipoprotein E; IL: Interleukin; TNFa: tumor necrosis factor alpha; NF-kB: nuclear transcription factor kB; LPS: lipopolysaccharide; CUMS: chronic unpredicted mild stress; AF: atrial fibrillation; RCT: randomized controlled trial, CAD: coronary artery disease; PAD: peripheral artery disease.

4.2. Apixaban

The current literature does not offer many studies testing the anti-inflammatory effects of apixaban (Table 2). Two in vitro studies demonstrated that cell culture incubation with apixaban limited expression of MCP-1 and adhesion molecules. [88,89] Additionally, in the model replicating the uremic-induced endothelial dysfunction, apixaban mitigated platelet adhesion to extracellular matrix and reactive oxygen species production. [89] Despite little preclinical evidence, a recent longitudinal study on 44 patients with acute cardioembolic ischemic stroke showed lower levels of IL-6 among those in the apixaban group [90].

4.3. Edoxaban

Only a few studies assessed the anti-inflammatory properties of edoxaban [91,92]. Although some investigations reported pleiotropic effects of edoxaban such as ROS reduction, anti-angiogenesis, and a decrease in the atherosclerotic plaque burden [93], we chose to report only studies focused on crucial inflammatory molecules (Table 3). In Fang et al. [91], randomized male wild-type mice were given edoxaban or a vehicle diet. Mice in the treatment group exhibited lower inflammatory biomarker levels. Additionally, HK2 cells exposed to edoxaban had suppressed NF-kB expression. Candido et al. [92] examined hospitalized patients with DVT. Participants initially received LWMH according to the guidelines and then were treated with either edoxaban or dabigatran. The 20 patients with DVT demonstrated a lower IL-6 expression when compared to the controls.

4.4. Dabigatran

A variety of preclinical studies highlighted the anti-inflammatory effects of dabigatran (Table 4). Hypoxia-exposed rat brain cells subjected to dabigatran showed a blunted inflammatory response with reduced expression of both IL-6 and MCP-1. A simultaneous analysis of brain sections from a transgenic mouse model of neurodegenerative disease yielded similar results [94]. Reduction of TNF-a and IL-1 was consistent across different studies evaluating the effectiveness of dabigatran in models of liver fibrosis, [95] tubulointerstitial fibrosis [96], and acute myocardial infarction [97]. Moreover, dabigatran-ameliorated diabetes-induced endothelial dysfunction fixed the expression of adhesion molecules [98]. Finally, the RIVAL-AF study compared anti-inflammatory effects of dabigatran and rivaroxaban in Japanese patients with non-valvular AF [99]. Dabigatran-treated patients had reduced levels of IL-6. Interestingly, although evaluating DOACs’ anti-inflammatory properties in different clinical scenarios, all DOAC clinical studies reported in this review showed a decreased expression of IL-6, suggesting a potential class effect.

5. Anticancer Effects of DOACs

5.1. Dabigatran

Most of the anticancer effects have been observed with dabigatran, paradoxically the only DOAC not yet recommended by guidelines in cancer patients (Table 5). A previous study clarified that several classes of proteases, including thrombin, work by directing cell activities through PARs [100]. Tumor initiation and growth, angiogenesis and metastatic spread are all influenced by thrombin.
An in vitro study, by DeFeo et al. [101], firstly described the anticancer properties of DOACs, showing that dabigatran decreased the spread of MDA-MB-231 breast cancer cells; this finding was also confirmed in vivo. Moreover, the authors explored potential antimetastatic effects of dabigatran, reporting a reduction in liver micrometastases when 4T1-cell-injected BALB/c mice were treated with dabigatran.
Alexander et al. evaluated the synergism of dabigatran either with cyclophosphamide or cisplatin [102,103]. Dabigatran alone considerably decreased circulating TF microparticles, even if the tumor suppression required co-administration with cyclophosphamide [102]. In a model of murine ovarian cancer, the co-treatment of dabigatran and cisplatin successfully reduced tumor growth and ascites [103]. Both studies [102,103] showed a reduction in the levels of proinflammatory cytokines and immunosuppressive cells with an established role in tumor promotion.
The potentiation of anticancer effects of common chemotherapeutic agents by dabigatran was shown not only for alkylating agents but also for antimetabolites. Indeed, in a murine model of pancreatic cancer, Shi et al. [105] found an increased gemcitabine cytotoxicity when dabigatran was co-administered. Finally, Vianello et al. [104] provided further mechanistic insights. Dabigatran prevented upregulation of cyclin D1 and proangiogenic factors in both glioblastoma and breast cancer cells; in addition, levels of p27 were restored.

5.2. Apixaban

Three in vitro studies explored the anticancer effects of apixaban [108,109,110] (Table 5). Guasti et al. [108] showed that the addition of apixaban to cancer cell cultures, i.e., epithelial cells of ovarian adenocarcinoma (OVCAR3), breast ductal cancer (MDA-MB 23), and colorectal adenocarcinoma (CaCO-2), significantly reduced proliferation and increased mortality of cancer cells through apoptosis in several cancer cell lines, except for the epithelial cells of prostate adenocarcinoma (LNCaP). No effect was noted on histiocytic lymphoma cells (U937). Conversely, cell vitality was lower for all cell lines, both solid and hematological. However, these effects are related to apixaban doses higher than those used clinically. Unfortunately, high doses of apixaban caused cytotoxic effects in endocervical cancer cells (HeLa) [109]. Featherby et al. [110] demonstrated an apixaban-mediated inhibition of proteolytic activity of FVII-TF complex; this effect prevents PAR-2 activation and subsequent release of TF-microvesicles, limiting proliferation.

5.3. Rivaroxaban

Besides conventional anticancer effects, such as the cytotoxic and antiproliferative effects investigated for other DOACs, rivaroxaban has proven antiangiogenic properties [106] and interesting effects on antitumor immunity [107] (Table 5).
In mice injected with T241 fibrosarcoma cells, Graf et al. [107] showed a reduction in tumor growth and metastases, suggesting that rivaroxaban promotes antitumor immunity by enhancing infiltration of dendritic cells and cytotoxic T cells at the tumor site. The combination of rivaroxaban and immunotherapy provided a synergistic effect, further limiting tumor growth compared to monotherapy. In particular, rivaroxaban worked on tumor microenvironment promoting CD103 + F4/80 − CCR7 + DC and GrB + cytotoxic CD8 + T cells. Their expression has been linked with better outcomes in cancer patients [112,113]. Moreover, immune-escape pathways supported by macrophages were blunted, as shown by a marked reduced expression in key markers (Mrc1, CD204).

5.4. Edoxaban

Only one study investigated the potential anticancer effects of edoxaban [111] (Table 5). In the first phase, all DOACs were studied in BALB/c mice injected with Colon26 cells. Since edoxaban exerted a greater suppression of tumor growth, it was chosen for further analysis. Plasmatic levels of prothrombotic factors such as TF, PAI-1, and MMP-2 were reduced. Additionally, the transcription of STAT3 and cyclin D1 was lowered, while p53 expression was upregulated.

6. Anti-inflammatory and Anticancer Effects of Heparins

Heparins are polysaccharides, belonging to the glycosaminoglycan (GAG) family, with anticoagulant effect due to their ability to bind antithrombin III (ATIII). Following the ATIII linkage, heparins increase the inhibitory effect of ATIII towards thrombin and factor X. Moreover, heparins reduce the prothrombotic state by inducing the cells to release tissue factor pathway inhibitor (TFPI), which exhibits both antimetastatic and antiangiogenic properties.
LMWHs are derived from chemical/enzymatic depolymerization of unfractionated heparin (UFH) and exhibit their anticoagulant function by activating the antithrombin and inhibiting the activation of factor X. LMWHs show more predictable functions and fewer adverse effects compared to UFH.
Besides the well-known anticoagulation effects, heparins seem to be involved in the regulation of the inflammation process, acting on neutrophil migration, complement activation, and cytokine production. The potential anticancer effects of heparins may be attributed to the reduction of prothrombotic state associated to malignancies. Moreover, a protective effect related to inhibiting of cancer progression by interfering with the metastatic process has been described [114,115,116].
Neoplastic cells present an altered expression of adhesion proteins and highly exceed proliferation rate when compared to normal cells. The augmented expression of selectin ligands may increase the metastatic process. Heparins bind to P-selectin, expressed by the platelets, preventing the interaction between platelets and neoplastic cells and reducing the probability of metastasis.
In a mouse model of colon carcinoma and melanoma, Hottlsler et al. showed a lower rate of metastases after heparin treatment. No effect of heparins was observed in mice deficient in P- and L-selectin, hence demonstrating the selectin-dependent function of heparin. [117] Heparin also reduced melanoma metastases in lungs by binding P-selectin expressed on endothelial cells [118] (Table 6). Tinzaparin seems to be the most effective in reducing metastatic spreading by the inhibition of P-selectin.
Growth factors are involved in neoplastic cell proliferation and angiogenesis. GAG mediates the signaling of growth factors and prevents their degradation. Heparins act as a bridge between growth factors and their receptors since they interact with both of them. Exogenous heparins, competing with GAG in the linkage to growth factors, may inhibit fibrosis and angiogenesis. However, only heparins with less than eight sugar units inhibit the VEGF/VEGF receptor interaction; in contrast, huge molecules seem to potentiate the tissue factor signaling, leading to a fast progression of cancer [116,117,118]. Other antiangiogenic potential effects of heparins may be related to the downregulation of factor twist and microRNA-10b (miR-10b), which are involved in cell migration and angiogenesis [122]. They also interfere with heparanase, an endoglycosidase overexpressed in cancer that cleaves heparan sulfate (HS), contributing to extracellular matrix remodeling and the spread of cancer cells [114,115,116,117,118,122,123].
Moreover, heparins showed a pro-apoptotic effect in peripheral blood leukocytes. In nasopharyngeal carcinoma cell line CNE2, heparin increased c-myc expression and bax/bcl2 ratio, both proteins involved in apoptosis regulation. [121]
In two in vitro studies, heparins increased sensitivity to chemotherapy and decreased the chemoresistance [119,120]. Enoxaparin plus gefitinib showed a more significant effect in reducing lung cancer volume and cell migration than gefitinib arm alone; these findings could be justified by a suppression of DOCK1 and AKT pathway, involved in cell migration and proliferation [119]. Moreover, heparins restored responsiveness to cisplatin in cisplatin-resistant ovarian cancer cells [120].
In a meta-analysis including nine studies with 2185 patients with lung cancer, the use of anticoagulant (VKA and subcutaneous heparin) showed a survival benefit and also prolonged life expectancy, especially for those with small-cell lung cancer (SCLC), even if anticoagulation was not indicated [124]. In an RCT by Ek et al. including 380 patients, the addition of enoxaparin at a supraprophylactic dose (1 mg/kg) to the standard treatment did not improve the overall survival, despite resulting in a significant reduction in VTE incidence. These results provides strong support against a more general use of LMWH as a tumor-inhibiting agent and underlines the need for risk biomarkers to guide clinicians in tailoring individualized LMWH treatment [86].

7. Clinical Implications

Several experimental in vitro and in vivo studies demonstrated the anti-inflammatory and anticancer effects of DOACs and LWMHs; however, to date, only the anti-inflammatory properties have been shown in studies involving humans. All currently approved DOACs, except dabigatran, are recommended as alternatives to LWMHs in the management of cancer-related VTE. Before starting anticoagulation therapy, a careful cardio-oncologic evaluation is required, as patients with gastrointestinal and genitourinary malignancy showed an increased risk of major bleeding when they were on DOACs therapy. A patient-centered approach based on the clinical characteristics and biomarker evaluation may help clinicians to choose the optimal anticoagulation strategy in patients with malignancy. In the era of precision medicine, the pleiotropic effects of anticoagulants are of significant interest and should be considered alongside drug–drug interactions and benefit/risk balance of drugs. Unfortunately, clinical data for direct anticancer effects are still lacking [125,126]. The “Thrombin Inhibition Preoperatively (TIP) in early breast cancer” is a clinical trial aiming to determine whether 14 days of a preoperative oral factor Xa inhibitor (rivaroxaban) in estrogen-receptor-negative early breast cancer patients results in inhibition of tumor proliferation determined by a reduction in tumor Ki67 from baseline (pre-treatment) to 14 days post treatment start (at the time of surgical excision).

8. Conclusions

Inflammation is a common pathway of thrombotic diseases and cancer. Several studies, both in vitro and in vivo, showed “pleiotropic” anti-inflammatory and anticancer effects of DOACs and LMWHs in cellular and animal models. However, to date, only the anti-inflammatory properties have been shown in clinical studies. Further studies are necessary to assess if this experimental evidence may have an impact on patients’ prognosis and overall survival. The use of DOACs as “adjuvant” treatment to chemotherapy, regardless of the patient’s thromboembolic risk, is an intriguing scenario that should be explored with further randomized clinical trials.

Author Contributions

Conceptualization: L.F. and V.R.; writing—original draft preparation: L.F., V.T., N.N., D.C., G.C., A.M., A.C. and V.R.; methodology: L.F., G.C., A.G. and V.R.; writing—review and editing: A.G., M.T., A.M., A.D. and V.R.; data curation: L.F., G.C., A.M., M.T., A.D. and V.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mulder, F.I.; Horváth-Puhó, E.; Van Es, N.; Van Laarhoven, H.W.M.; Pedersen, L.; Moik, F.; Ay, C.; Büller, H.R.; Sørensen, H.T. Venous Thromboembolism in Cancer Patients: A Population-Based Cohort Study. Blood 2021, 137, 1959–1969. [Google Scholar] [CrossRef]
  2. Khorana, A.A.; Francis, C.W.; Culakova, E.; Kuderer, N.M.; Lyman, G.H. Thromboembolism Is a Leading Cause of Death in Cancer Patients Receiving Outpatient Chemotherapy. J. Thromb. Haemost. 2007, 5, 632–634. [Google Scholar] [CrossRef] [PubMed]
  3. Lee, A.Y.Y.; Levine, M.N.; Baker, R.I.; Bowden, C.; Kakkar, A.K.; Prins, M.; Rickles, F.R.; Julian, J.A.; Haley, S.; Kovacs, M.J.; et al. Low-Molecular-Weight Heparin versus a Coumarin for the Prevention of Recurrent Venous Thromboembolism in Patients with Cancer. N. Engl. J. Med. 2003, 349, 146–153. [Google Scholar] [CrossRef] [PubMed]
  4. Kirkilesis, G.I.; Kakkos, S.K.; Tsolakis, I.A. Editor’s Choice—A Systematic Review and Meta-Analysis of the Efficacy and Safety of Anticoagulation in the Treatment of Venous Thromboembolism in Patients with Cancer. Eur. J. Vasc. Endovasc. Surg. 2019, 57, 685–701. [Google Scholar] [CrossRef]
  5. Khorana, A.A.; McCrae, K.R.; Milentijevic, D.; Fortier, J.; Nelson, W.W.; Laliberté, F.; Crivera, C.; Lefebvre, P.; Yannicelli, D.; Schein, J. Current Practice Patterns and Patient Persistence with Anticoagulant Treatments for Cancer-associated Thrombosis. Res. Pract. Thromb. Haemost. 2017, 1, 14–22. [Google Scholar] [CrossRef]
  6. Ortel, T.L.; Neumann, I.; Ageno, W.; Beyth, R.; Clark, N.P.; Cuker, A.; Hutten, B.A.; Jaff, M.R.; Manja, V.; Schulman, S.; et al. American Society of Hematology 2020 Guidelines for Management of Venous Thromboembolism: Treatment of Deep Vein Thrombosis and Pulmonary Embolism. Blood Adv. 2020, 4, 4693–4738. [Google Scholar] [CrossRef] [PubMed]
  7. Potpara, T.; Dagres, N.; Arbelo, E.; Bax, J.J.; Boriani, G.; Castella, M.; Dan, G.-A.; Dilaveris, P.E.; Fauchier, L.; Filippatos, G.; et al. The Task Force for the Diagnosis and Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Eur. Heart J. 2021, 42, 373–498. [Google Scholar]
  8. Ruff, C.T.; Giugliano, R.P.; Braunwald, E.; Hoffman, E.B.; Deenadayalu, N.; Ezekowitz, M.D.; Camm, A.J.; Weitz, J.I.; Lewis, B.S.; Parkhomenko, A.; et al. Comparison of the Efficacy and Safety of New Oral Anticoagulants with Warfarin in Patients with Atrial Fibrillation: A Meta-Analysis of Randomised Trials. Lancet 2014, 383, 955–962. [Google Scholar] [CrossRef] [PubMed]
  9. 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]
  10. Russo, V.; Cattaneo, D.; Giannetti, L.; Bottino, R.; Laezza, N.; Atripaldi, U.; Clementi, E. Pharmacokinetics of Direct Oral Anticoagulants in Patients with Atrial Fibrillation and Extreme Obesity. Clin. Ther. 2021, 43, e255–e263. [Google Scholar] [CrossRef]
  11. Russo, V.; Rago, A.; Papa, A.; Meo, F.; Attena, E.; Golino, P.; D’Onofrio, A.; Nigro, G. Use of Non–Vitamin K Antagonist Oral Anticoagulants in Atrial Fibrillation Patients with Malignancy: Clinical Practice Experience in a Single Institution and Literature Review. Semin. Thromb. Hemost. 2018, 44, 370–376. [Google Scholar] [CrossRef]
  12. Russo, V.; Bottino, R.; Rago, A.; Micco, P.; D’Onofrio, A.; Liccardo, B.; Golino, P.; Nigro, G. Atrial Fibrillation and Malignancy: The Clinical Performance of Non–Vitamin K Oral Anticoagulants—A Systematic Review. Semin. Thromb. Hemost. 2019, 45, 205–214. [Google Scholar] [CrossRef]
  13. Melillo, E.; Carbone, A.; Rago, A.; Papa, A.A.; Onofrio, A.D.; Nigro, G.; Golino, P.; Russo, V. Update on Direct Oral Anticoagulants in Atrial Fibrillation Patients Undergoing Cardiac Interventional Procedures: From Clinical Trials to Real-World Evidence. J. Cardiovasc. Pharmacol. 2020, 75, 185–199. [Google Scholar] [CrossRef]
  14. Carbone, A.; Santelli, F.; Bottino, R.; Attena, E.; Mazzone, C.; Parisi, V.; D’Andrea, A.; Golino, P.; Nigro, G.; Russo, V. Prevalence and Clinical Predictors of Inappropriate Direct Oral Anticoagulant Dosage in Octagenarians with Atrial Fibrillation. Eur. J. Clin. Pharmacol. 2022, 78, 879–886. [Google Scholar] [CrossRef]
  15. Carbone, A.; Bottino, R.; Attena, E.; Parisi, V.; Conte, M.; D’Andrea, A.; Imbalzano, E.; Golino, P.; Russo, V. Clinical Impact of Oral Anticoagulation among Octogenarians with Atrial Fibrillation and Anaemia. J. Thromb. Thrombolysis 2022, 55, 222–227. [Google Scholar] [CrossRef]
  16. Russo, V.; Attena, E.; Di Maio, M.; Carbone, A.; Parisi, V.; Rago, A.; Grieco, F.V.; Buonauro, A.; Golino, P.; Nigro, G. Non-vitamin K vs Vitamin K Oral Anticoagulants in Patients Aged > 80 Year with Atrial Fibrillation and Low Body Weight. Eur. J. Clin. Investig 2020, 50, e13335. [Google Scholar] [CrossRef] [PubMed]
  17. Russo, V.; Attena, E.; Di Maio, M.; Mazzone, C.; Carbone, A.; Parisi, V.; Rago, A.; D’Onofrio, A.; Golino, P.; Nigro, G. Clinical Profile of Direct Oral Anticoagulants versus Vitamin K Anticoagulants in Octogenarians with Atrial Fibrillation: A Multicentre Propensity Score Matched Real-World Cohort Study. J. Thromb. Thrombolysis 2020, 49, 42–53. [Google Scholar] [CrossRef] [PubMed]
  18. Russo, V.; Attena, E.; Mazzone, C.; Esposito, F.; Parisi, V.; Bancone, C.; Rago, A.; Nigro, G.; Sangiuolo, R.; D’ Onofrio, A. Nonvitamin K Antagonist Oral Anticoagulants Use in Patients with Atrial Fibrillation and Bioprosthetic Heart Valves/Prior Surgical Valve Repair: A Multicenter Clinical Practice Experience. Semin. Thromb. Hemost. 2018, 44, 364–369. [Google Scholar] [CrossRef] [PubMed]
  19. Russo, V.; Carbone, A.; Attena, E.; Rago, A.; Mazzone, C.; Proietti, R.; Parisi, V.; Scotti, A.; Nigro, G.; Golino, P.; et al. Clinical Benefit of Direct Oral Anticoagulants Versus Vitamin K Antagonists in Patients with Atrial Fibrillation and Bioprosthetic Heart Valves. Clin. Ther. 2019, 41, 2549–2557. [Google Scholar] [CrossRef]
  20. Melillo, E.; Rago, A.; Proietti, R.; Attena, E.; Carrella, M.; Golino, P.; D’Onofrio, A.; Nigro, G.; Russo, V. Atrial Fibrillation and Mitral Regurgitation: Clinical Performance of Direct Oral Anticoagulants in a Real-World Setting. J. Cardiovasc. Pharmacol. Ther. 2020, 25, 564–569. [Google Scholar] [CrossRef]
  21. Russo, V.; Attena, E.; Baroni, M.; Trotta, R.; Manu, M.C.; Kirchhof, P.; De Caterina, R. Clinical Performance of Oral Anticoagulants in Elderly with Atrial Fibrillation and Low Body Weight: Insight into Italian Cohort of PREFER-AF and PREFER-AF Prolongation Registries. J. Clin. Med. 2022, 11, 3751. [Google Scholar] [CrossRef]
  22. Carbone, A.; Bottino, R.; D’Andrea, A.; Russo, V. Direct Oral Anticoagulants for Stroke Prevention in Special Populations: Beyond the Clinical Trials. Biomedicines 2023, 11, 131. [Google Scholar] [CrossRef]
  23. Russo, V.; Carbone, A.; Rago, A.; Golino, P.; Nigro, G. Direct Oral Anticoagulants in Octogenarians with Atrial Fibrillation: It Is Never Too Late. J. Cardiovasc. Pharmacol. 2019, 73, 207–214. [Google Scholar] [CrossRef] [PubMed]
  24. Raskob, G.E.; Van Es, N.; Verhamme, P.; Carrier, M.; Di Nisio, M.; Garcia, D.; Grosso, M.A.; Kakkar, A.K.; Kovacs, M.J.; Mercuri, M.F.; et al. Edoxaban for the Treatment of Cancer-Associated Venous Thromboembolism. N. Engl. J. Med. 2018, 378, 615–624. [Google Scholar] [CrossRef] [PubMed]
  25. Young, A.M.; Marshall, A.; Thirlwall, J.; Chapman, O.; Lokare, A.; Hill, C.; Hale, D.; Dunn, J.A.; Lyman, G.H.; Hutchinson, C.; et al. Comparison of an Oral Factor Xa Inhibitor with Low Molecular Weight Heparin in Patients With Cancer With Venous Thromboembolism: Results of a Randomized Trial (SELECT-D). JCO 2018, 36, 2017–2023. [Google Scholar] [CrossRef]
  26. McBane, R.D.; Wysokinski, W.E.; Le-Rademacher, J.G.; Zemla, T.; Ashrani, A.; Tafur, A.; Perepu, U.; Anderson, D.; Gundabolu, K.; Kuzma, C.; et al. Apixaban and Dalteparin in Active Malignancy-associated Venous Thromboembolism: The ADAM VTE Trial. J. Thromb. Haemost. 2020, 18, 411–421. [Google Scholar] [CrossRef]
  27. Agnelli, G.; Becattini, C.; Meyer, G.; Muñoz, A.; Huisman, M.V.; Connors, J.M.; Cohen, A.; Bauersachs, R.; Brenner, B.; Torbicki, A.; et al. Apixaban for the Treatment of Venous Thromboembolism Associated with Cancer. N. Engl. J. Med. 2020, 382, 1599–1607. [Google Scholar] [CrossRef]
  28. Planquette, B.; Bertoletti, L.; Charles-Nelson, A.; Laporte, S.; Grange, C.; Mahé, I.; Pernod, G.; Elias, A.; Couturaud, F.; Falvo, N.; et al. Rivaroxaban vs Dalteparin in Cancer-Associated Thromboembolism. Chest 2022, 161, 781–790. [Google Scholar] [CrossRef] [PubMed]
  29. Key, N.S.; Khorana, A.A.; Kuderer, N.M.; Bohlke, K.; Lee, A.Y.Y.; Arcelus, J.I.; Wong, S.L.; Balaban, E.P.; Flowers, C.R.; Francis, C.W.; et al. Venous Thromboembolism Prophylaxis and Treatment in Patients with Cancer: ASCO Clinical Practice Guideline Update. JCO 2020, 38, 496–520. [Google Scholar] [CrossRef] [PubMed]
  30. Lyman, G.H.; Carrier, M.; Ay, C.; Di Nisio, M.; Hicks, L.K.; Khorana, A.A.; Leavitt, A.D.; Lee, A.Y.Y.; Macbeth, F.; Morgan, R.L.; et al. American Society of Hematology 2021 Guidelines for Management of Venous Thromboembolism: Prevention and Treatment in Patients with Cancer. Blood Adv. 2021, 5, 927–974. [Google Scholar] [CrossRef]
  31. Rossi, J.-F.; Lu, Z.Y.; Massart, C.; Levon, K. Dynamic Immune/Inflammation Precision Medicine: The Good and the Bad Inflammation in Infection and Cancer. Front. Immunol. 2021, 12, 595722. [Google Scholar] [CrossRef]
  32. Balkwill, F.; Mantovani, A. Inflammation and Cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  33. Rogovskii, V. Modulation of Inflammation-Induced Tolerance in Cancer. Front. Immunol. 2020, 11, 1180. [Google Scholar] [CrossRef]
  34. Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and Tumor Progression: Signaling Pathways and Targeted Intervention. Sig. Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [CrossRef] [PubMed]
  35. Murata, M. Inflammation and Cancer. Environ. Health Prev. Med. 2018, 23, 50. [Google Scholar] [CrossRef]
  36. Xiao, L.; Li, X.; Cao, P.; Fei, W.; Zhou, H.; Tang, N.; Liu, Y. Interleukin-6 Mediated Inflammasome Activation Promotes Oral Squamous Cell Carcinoma Progression via JAK2/STAT3/Sox4/NLRP3 Signaling Pathway. J. Exp. Clin. Cancer Res. 2022, 41, 166. [Google Scholar] [CrossRef]
  37. Crusz, S.M.; Balkwill, F.R. Inflammation and Cancer: Advances and New Agents. Nat. Rev. Clin. Oncol. 2015, 12, 584–596. [Google Scholar] [CrossRef]
  38. Wang, L.; Lan, J.; Tang, J.; Luo, N. MCP-1 Targeting: Shutting off an Engine for Tumor Development (Review). Oncol. Lett. 2021, 23, 26. [Google Scholar] [CrossRef] [PubMed]
  39. Sun, S.-C. The Non-Canonical NF-ΚB Pathway in Immunity and Inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef] [PubMed]
  40. Tegowski, M.; Baldwin, A. Noncanonical NF-ΚB in Cancer. Biomedicines 2018, 6, 66. [Google Scholar] [CrossRef]
  41. Franco, A.T.; Corken, A.; Ware, J. Platelets at the Interface of Thrombosis, Inflammation, and Cancer. Blood 2015, 126, 582–588. [Google Scholar] [CrossRef]
  42. Stark, K.; Massberg, S. Interplay between Inflammation and Thrombosis in Cardiovascular Pathology. Nat. Rev. Cardiol. 2021, 18, 666–682. [Google Scholar] [CrossRef]
  43. Vazquez-Garza, E.; Jerjes-Sanchez, C.; Navarrete, A.; Joya-Harrison, J.; Rodriguez, D. Venous Thromboembolism: Thrombosis, Inflammation, and Immunothrombosis for Clinicians. J. Thromb. Thrombolysis 2017, 44, 377–385. [Google Scholar] [CrossRef] [PubMed]
  44. Hickey, M.J.; Kubes, P. Intravascular Immunity: The Host–Pathogen Encounter in Blood Vessels. Nat. Rev. Immunol. 2009, 9, 364–375. [Google Scholar] [CrossRef] [PubMed]
  45. Battinelli, E.M.; Hartwig, J.H.; Italiano, J.E. Delivering New Insight into the Biology of Megakaryopoiesis and Thrombopoiesis. Curr. Opin. Hematol. 2007, 14, 419–426. [Google Scholar] [CrossRef] [PubMed]
  46. Esmon, C.T. The Interactions between Inflammation and Coagulation. Br. J. Haematol. 2005, 131, 417–430. [Google Scholar] [CrossRef] [PubMed]
  47. Beaulieu, L.M.; Lin, E.; Mick, E.; Koupenova, M.; Weinberg, E.O.; Kramer, C.D.; Genco, C.A.; Tanriverdi, K.; Larson, M.G.; Benjamin, E.J.; et al. Interleukin 1 Receptor 1 and Interleukin 1β Regulate Megakaryocyte Maturation, Platelet Activation, and Transcript Profile During Inflammation in Mice and Humans. ATVB 2014, 34, 552–564. [Google Scholar] [CrossRef]
  48. Cognasse, F.; Duchez, A.C.; Audoux, E.; Ebermeyer, T.; Arthaud, C.A.; Prier, A.; Eyraud, M.A.; Mismetti, P.; Garraud, O.; Bertoletti, L.; et al. Platelets as Key Factors in Inflammation: Focus on CD40L/CD40. Front. Immunol. 2022, 13, 825892. [Google Scholar] [CrossRef]
  49. Esmon, C.T. The Regulation of Natural Anticoagulant Pathways. Science 1987, 235, 1348–1352. [Google Scholar] [CrossRef]
  50. Minami, T.; Sugiyama, A.; Wu, S.-Q.; Abid, R.; Kodama, T.; Aird, W.C. Thrombin and Phenotypic Modulation of the Endothelium. ATVB 2004, 24, 41–53. [Google Scholar] [CrossRef]
  51. Ossovskaya, V.S.; Bunnett, N.W. Protease-Activated Receptors: Contribution to Physiology and Disease. Physiol. Rev. 2004, 84, 579–621. [Google Scholar] [CrossRef] [PubMed]
  52. Xu, W.; Andersen, H.; Whitmore, T.E.; Presnell, S.R.; Yee, D.P.; Ching, A.; Gilbert, T.; Davie, E.W.; Foster, D.C. Cloning and Characterization of Human Protease-Activated Receptor 4. Proc. Natl. Acad. Sci. USA 1998, 95, 6642–6646. [Google Scholar] [CrossRef] [PubMed]
  53. Bae, J.-S.; Yang, L.; Rezaie, A.R. Factor X/Xa Elicits Protective Signaling Responses in Endothelial Cells Directly via PAR-2 and Indirectly via Endothelial Protein C Receptor-Dependent Recruitment of PAR-1. J. Biol. Chem. 2010, 285, 34803–34812. [Google Scholar] [CrossRef] [PubMed]
  54. Riewald, M.; Kravchenko, V.V.; Petrovan, R.J.; O’Brien, P.J.; Brass, L.F.; Ulevitch, R.J.; Ruf, W. Gene Induction by Coagulation Factor Xa Is Mediated by Activation of Protease-Activated Receptor 1. Blood 2001, 97, 3109–3116. [Google Scholar] [CrossRef]
  55. Jiang, R.; Wang, N.-P.; Tanaka, K.A.; Levy, J.H.; Guyton, R.A.; Zhao, Z.-Q.; Vinten-Johansen, J. Factor Xa Induces Tissue Factor Expression in Endothelial Cells by P44/42 MAPK and NF-ΚB-Dependent Pathways. J. Surg. Res. 2011, 169, 319–327. [Google Scholar] [CrossRef]
  56. Busch, G.; Seitz, I.; Steppich, B.; Hess, S.; Eckl, R.; Schömig, A.; Ott, I. Coagulation Factor Xa Stimulates Interleukin-8 Release in Endothelial Cells and Mononuclear Leukocytes: Implications in Acute Myocardial Infarction. ATVB 2005, 25, 461–466. [Google Scholar] [CrossRef]
  57. Chen, D.; Carpenter, A.; Abrahams, J.; Chambers, R.C.; Lechler, R.I.; McVey, J.H.; Dorling, A. Protease-Activated Receptor 1 Activation Is Necessary for Monocyte Chemoattractant Protein 1–Dependent Leukocyte Recruitment in Vivo. J. Exp. Med. 2008, 205, 1739–1746. [Google Scholar] [CrossRef]
  58. Sébert, M.; Sola-Tapias, N.; Mas, E.; Barreau, F.; Ferrand, A. Protease-Activated Receptors in the Intestine: Focus on Inflammation and Cancer. Front. Endocrinol. 2019, 10, 717. [Google Scholar] [CrossRef]
  59. Scotton, C.J.; Krupiczojc, M.A.; Königshoff, M.; Mercer, P.F.; Lee, Y.C.G.; Kaminski, N.; Morser, J.; Post, J.M.; Maher, T.M.; Nicholson, A.G.; et al. Increased Local Expression of Coagulation Factor X Contributes to the Fibrotic Response in Human and Murine Lung Injury. J. Clin. Investig. 2009, 119, 2550–2563. [Google Scholar] [CrossRef] [PubMed]
  60. Borensztajn, K.; Stiekema, J.; Nijmeijer, S.; Reitsma, P.H.; Peppelenbosch, M.P.; Spek, C.A. Factor Xa Stimulates Proinflammatory and Profibrotic Responses in Fibroblasts via Protease-Activated Receptor-2 Activation. Am. J. Pathol. 2008, 172, 309–320. [Google Scholar] [CrossRef]
  61. Heuberger, D.M.; Franchini, A.G.; Madon, J.; Schuepbach, R.A. Thrombin Cleaves and Activates the Protease-Activated Receptor 2 Dependent on Thrombomodulin Co-Receptor Availability. Thromb. Res. 2019, 177, 91–101. [Google Scholar] [CrossRef] [PubMed]
  62. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on Cardio-Oncology Developed in Collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef]
  63. Efficacy and Safety of Enoxaparin versus Unfractionated Heparin for Prevention of Deep Vein Thrombosis in Elective Cancer Surgery: A Double-Blind Randomized Multicentre Trial with Venographic Assessment: PREVENTION OF DEEP VEIN THROMBOSIS IN CANCER SURGERY. Br. J. Surg. 1997, 84, 1099–1103. [CrossRef]
  64. Mulder, F.I.; Candeloro, M.; Kamphuisen, P.W.; Di Nisio, M.; Bossuyt, P.M.; Guman, N.; Smit, K.; Büller, H.R.; Van Es, N. The Khorana Score for Prediction of Venous Thromboembolism in Cancer Patients: A Systematic Review and Meta-Analysis. Haematologica 2019, 104, 1277–1287. [Google Scholar] [CrossRef]
  65. Gerotziafas, G.T.; Taher, A.; Abdel-Razeq, H.; AboElnazar, E.; Spyropoulos, A.C.; El Shemmari, S.; Larsen, A.K.; Elalamy, I.; on behalf of the COMPASS–CAT Working Group. A Predictive Score for Thrombosis Associated with Breast, Colorectal, Lung, or Ovarian Cancer: The Prospective COMPASS–Cancer-Associated Thrombosis Study. Oncologist 2017, 22, 1222–1231. [Google Scholar] [CrossRef]
  66. Carrier, M.; Abou-Nassar, K.; Mallick, R.; Tagalakis, V.; Shivakumar, S.; Schattner, A.; Kuruvilla, P.; Hill, D.; Spadafora, S.; Marquis, K.; et al. Apixaban to Prevent Venous Thromboembolism in Patients with Cancer. N. Engl. J. Med. 2019, 380, 711–719. [Google Scholar] [CrossRef] [PubMed]
  67. Bauersachs, R.; Khorana, A.A.; Lee, A.Y.Y.; Soff, G. Cancer-associated Venous Thromboembolism: Treatment and Prevention with Rivaroxaban. Res. Pract. Thromb. Haemost. 2020, 4, 532–549. [Google Scholar] [CrossRef]
  68. January, C.T.; Wann, L.S.; Calkins, H.; Chen, L.Y.; Cigarroa, J.E.; Cleveland, J.C.; Ellinor, P.T.; Ezekowitz, M.D.; Field, M.E.; Furie, K.L.; et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society in Collaboration With the Society of Thoracic Surgeons. Circulation 2019, 140, e125–e151. [Google Scholar] [CrossRef]
  69. Boriani, G.; Lee, G.; Parrini, I.; Lopez-Fernandez, T.; Lyon, A.R.; Suter, T.; Van Der Meer, P.; Cardinale, D.; Lancellotti, P.; Zamorano, J.L.; et al. Anticoagulation in Patients with Atrial Fibrillation and Active Cancer: An International Survey on Patient Management. Eur. J. Prev. Cardiol. 2021, 28, 611–621. [Google Scholar] [CrossRef]
  70. Falanga, A.; Ay, C.; Di Nisio, M.; Gerotziafas, G.; Jara-Palomares, L.; Langer, F.; Lecumberri, R.; Mandala, M.; Maraveyas, A.; Pabinger, I.; et al. Venous Thromboembolism in Cancer Patients: ESMO Clinical Practice Guideline. Ann. Oncol. 2023, 34, 452–467. [Google Scholar] [CrossRef]
  71. Farmakis, D.; Papakotoulas, P.; Angelopoulou, E.; Bischiniotis, T.; Giannakoulas, G.; Kliridis, P.; Richter, D.; Paraskevaidis, I. Anticoagulation for Atrial Fibrillation in Active Cancer (Review). Oncol. Lett. 2022, 23, 124. [Google Scholar] [CrossRef] [PubMed]
  72. Bansilal, S.; Bloomgarden, Z.; Halperin, J.L.; Hellkamp, A.S.; Lokhnygina, Y.; Patel, M.R.; Becker, R.C.; Breithardt, G.; Hacke, W.; Hankey, G.J.; et al. Efficacy and Safety of Rivaroxaban in Patients with Diabetes and Nonvalvular Atrial Fibrillation: The Rivaroxaban Once-Daily, Oral, Direct Factor Xa Inhibition Compared with Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation (ROCKET AF Trial). Am. Heart J. 2015, 170, 675–682.e8. [Google Scholar] [CrossRef] [PubMed]
  73. Guimarães, P.O.; Pokorney, S.D.; Lopes, R.D.; Wojdyla, D.M.; Gersh, B.J.; Giczewska, A.; Carnicelli, A.; Lewis, B.S.; Hanna, M.; Wallentin, L.; et al. Efficacy and Safety of Apixaban vs Warfarin in Patients with Atrial Fibrillation and Prior Bioprosthetic Valve Replacement or Valve Repair: Insights from the ARISTOTLE Trial. Clin. Cardiol. 2019, 42, 568–571. [Google Scholar] [CrossRef]
  74. Kato, E.T.; Giugliano, R.P.; Ruff, C.T.; Koretsune, Y.; Yamashita, T.; Kiss, R.G.; Nordio, F.; Murphy, S.A.; Kimura, T.; Jin, J.; et al. Efficacy and Safety of Edoxaban in Elderly Patients with Atrial Fibrillation in the ENGAGE AF–TIMI 48 Trial. J. Am. Heart Assoc. 2016, 5, e003432. [Google Scholar] [CrossRef]
  75. Bukowska, A.; Zacharias, I.; Weinert, S.; Skopp, K.; Hartmann, C.; Huth, C.; Goette, A. Coagulation Factor Xa Induces an Inflammatory Signalling by Activation of Protease-Activated Receptors in Human Atrial Tissue. Eur. J. Pharmacol. 2013, 718, 114–123. [Google Scholar] [CrossRef]
  76. Ishibashi, Y.; Matsui, T.; Ueda, S.; Fukami, K.; Yamagishi, S. Advanced Glycation End Products Potentiate Citrated Plasma-Evoked Oxidative and Inflammatory Reactions in Endothelial Cells by up-Regulating Protease-Activated Receptor-1 Expression. Cardiovasc. Diabetol. 2014, 13, 60. [Google Scholar] [CrossRef]
  77. Ishibashi, Y.; Matsui, T.; Fukami, K.; Ueda, S.; Okuda, S.; Yamagishi, S. Rivaroxaban Inhibits Oxidative and Inflammatory Reactions in Advanced Glycation End Product-Exposed Tubular Cells by Blocking Thrombin/Protease-Activated Receptor-2 System. Thromb. Res. 2015, 135, 770–773. [Google Scholar] [CrossRef]
  78. Ellinghaus, P.; Perzborn, E.; Hauenschild, P.; Gerdes, C.; Heitmeier, S.; Visser, M.; Summer, H.; Laux, V. Expression of Pro-Inflammatory Genes in Human Endothelial Cells: Comparison of Rivaroxaban and Dabigatran. Thromb. Res. 2016, 142, 44–51. [Google Scholar] [CrossRef] [PubMed]
  79. Zhou, Q.; Bea, F.; Preusch, M.; Wang, H.; Isermann, B.; Shahzad, K.; Katus, H.A.; Blessing, E. Evaluation of Plaque Stability of Advanced Atherosclerotic Lesions in Apo E-Deficient Mice after Treatment with the Oral Factor Xa Inhibitor Rivaroxaban. Mediat. Inflamm. 2011, 2011, 432080. [Google Scholar] [CrossRef] [PubMed]
  80. Hara, T.; Fukuda, D.; Tanaka, K.; Higashikuni, Y.; Hirata, Y.; Nishimoto, S.; Yagi, S.; Yamada, H.; Soeki, T.; Wakatsuki, T.; et al. Rivaroxaban, a Novel Oral Anticoagulant, Attenuates Atherosclerotic Plaque Progression and Destabilization in ApoE-Deficient Mice. Atherosclerosis 2015, 242, 639–646. [Google Scholar] [CrossRef]
  81. Imano, H.; Kato, R.; Tanikawa, S.; Yoshimura, F.; Nomura, A.; Ijiri, Y.; Yamaguchi, T.; Izumi, Y.; Yoshiyama, M.; Hayashi, T. Factor Xa Inhibition by Rivaroxaban Attenuates Cardiac Remodeling Due to Intermittent Hypoxia. J. Pharmacol. Sci. 2018, 137, 274–282. [Google Scholar] [CrossRef]
  82. Daci, A.; Da Dalt, L.; Alaj, R.; Shurdhiqi, S.; Neziri, B.; Ferizi, R.; Danilo Norata, G.; Krasniqi, S. Rivaroxaban Improves Vascular Response in LPS-Induced Acute Inflammation in Experimental Models. PLoS ONE 2020, 15, e0240669. [Google Scholar] [CrossRef] [PubMed]
  83. Abdelzaher, W.Y.; Mohammed, H.H.; Welson, N.N.; Batiha, G.E.-S.; Baty, R.S.; Abdel-Aziz, A.M. Rivaroxaban Modulates TLR4/Myd88/NF-Kβ Signaling Pathway in a Dose-Dependent Manner with Suppression of Oxidative Stress and Inflammation in an Experimental Model of Depression. Front. Pharmacol. 2021, 12, 715354. [Google Scholar] [CrossRef]
  84. Martins, G.L.; Duarte, R.C.F.; Vieira, É.L.M.; Rocha, N.P.; Figueiredo, E.L.; Silveira, F.R.; Caiaffa, J.R.S.; Lanna, R.P.; Carvalho, M.D.G.; Palotás, A.; et al. Comparison of Inflammatory Mediators in Patients with Atrial Fibrillation Using Warfarin or Rivaroxaban. Front. Cardiovasc. Med. 2020, 7, 114. [Google Scholar] [CrossRef] [PubMed]
  85. Kirchhof, P.; Ezekowitz, M.D.; Purmah, Y.; Schiffer, S.; Meng, I.L.; Camm, A.J.; Hohnloser, S.H.; Schulz, A.; Wosnitza, M.; Cappato, R. Effects of Rivaroxaban on Biomarkers of Coagulation and Inflammation: A Post Hoc Analysis of the X-VeRT Trial. TH Open 2020, 04, e20–e32. [Google Scholar] [CrossRef] [PubMed]
  86. Ek, L.; Gezelius, E.; Bergman, B.; Bendahl, P.O.; Anderson, H.; Sundberg, J.; Wallberg, M.; Falkmer, U.; Verma, S.; Belting, M. Swedish Lung Cancer Study Group (SLUSG). Randomized phase III trial of low-molecular-weight heparin enoxaparin in addition to standard treatment in small-cell lung cancer: The RASTEN trial. Ann. Oncol. 2018, 29, 398–404. [Google Scholar] [CrossRef] [PubMed]
  87. Russo, V.; Fabiani, D.; Leonardi, S.; Attena, E.; D’Alterio, G.; Cotticelli, C.; Rago, A.; Sarpa, S.; Maione, B.; D’Onofrio, A.; et al. Dual Pathway Inhibition with Rivaroxaban and Aspirin Reduces Inflammatory Biomarkers in Atherosclerosis. J. Cardiovasc. Pharmacol. 2023, 81, 129–133. [Google Scholar] [CrossRef]
  88. Ishibashi, Y.; Matsui, T.; Yamagishi, S. Apixaban Exerts Anti-Inflammatory Effects in Mesangial Cells by Blocking Thrombin/Protease-Activated Receptor-1 System. Thromb. Res. 2014, 134, 1365–1367. [Google Scholar] [CrossRef] [PubMed]
  89. Torramade-Moix, S.; Palomo, M.; Vera, M.; Jerez, D.; Moreno-Castaño, A.B.; Zafar, M.U.; Rovira, J.; Diekmann, F.; Garcia-Pagan, J.C.; Escolar, G.; et al. Apixaban Downregulates Endothelial Inflammatory and Prothrombotic Phenotype in an In Vitro Model of Endothelial Dysfunction in Uremia. Cardiovasc. Drugs Ther. 2021, 35, 521–532. [Google Scholar] [CrossRef]
  90. Nakase, T.; Moroi, J.; Ishikawa, T. Anti-inflammatory and Antiplatelet Effects of Non-vitamin K Antagonist Oral Anticoagulants in Acute Phase of Ischemic Stroke Patients. Clin. Transl. Med. 2018, 7, 2. [Google Scholar] [CrossRef]
  91. Fang, L.; Ohashi, K.; Ogawa, H.; Otaka, N.; Kawanishi, H.; Takikawa, T.; Ozaki, Y.; Takahara, K.; Tatsumi, M.; Takefuji, M.; et al. Factor Xa Inhibitor, Edoxaban Ameliorates Renal Injury after Subtotal Nephrectomy by Reducing Epithelial-mesenchymal Transition and Inflammatory Response. Physiol. Rep. 2022, 10, e15218. [Google Scholar] [CrossRef]
  92. Candido, S.; Lumera, G.; Barcellona, G.; Vetri, D.; Tumino, E.; Platania, I.; Frazzetto, E.; Privitera, G.; Incognito, C.; Gaudio, A.; et al. Direct Oral Anticoagulant Treatment of Deep Vein Thrombosis Reduces IL-6 Expression in Peripheral Mono-nuclear Blood Cells. Exp. Ther. Med. 2020, 20, 237. [Google Scholar] [CrossRef] [PubMed]
  93. Almengló, C.; Mosquera-Garrote, N.; González-Peteiro, M.; González-Juanatey, J.R.; Álvarez, E. Edoxaban’s Contribution to Key Endothelial Cell Functions. Biochem. Pharmacol. 2020, 178, 114063. [Google Scholar] [CrossRef] [PubMed]
  94. Tripathy, D.; Sanchez, A.; Yin, X.; Luo, J.; Martinez, J.; Grammas, P. Thrombin, a Mediator of Cerebrovascular Inflammation in AD and Hypoxia. Front. Aging Neurosci. 2013, 5, 19. [Google Scholar] [CrossRef]
  95. Mahmoud, N.I.; Messiha, B.A.S.; Salehc, I.G.; Abo-Saif, A.A.; Abdel-Bakky, M.S. Interruption of Platelets and Thrombin Function as a New Approach against Liver Fibrosis Induced Experimentally in Rats. Life Sci. 2019, 231, 116522. [Google Scholar] [CrossRef]
  96. Saifi, M.A.; Annaldas, S.; Godugu, C. A Direct Thrombin Inhibitor, Dabigatran Etexilate Protects from Renal Fibrosis by Inhibiting Protease Activated Receptor-1. Eur. J. Pharmacol. 2021, 893, 173838. [Google Scholar] [CrossRef]
  97. Song, K.; Wang, Y.; Sheng, J.; Ma, C.; Li, H. Effects of Dabigatran Regulates No-reflow Phenomenon in Acute Myocardial Infarction Mice through Anti-inflammatory and Anti-oxidative Activities and Connective Tissue Growth Factor Expression. Mol. Med. Rep. 2017, 18, 580–585. [Google Scholar] [CrossRef] [PubMed]
  98. Rahadian, A.; Fukuda, D.; Salim, H.M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Shimabukuro, M.; Sata, M. Thrombin Inhibition by Dabigatran Attenuates Endothelial Dysfunction in Diabetic Mice. Vasc. Pharmacol. 2020, 124, 106632. [Google Scholar] [CrossRef] [PubMed]
  99. Kikuchi, S.; Tsukahara, K.; Sakamaki, K.; Morita, Y.; Takamura, T.; Fukui, K.; Endo, T.; Shimizu, M.; Sawada, R.; Sugano, T.; et al. Comparison of Anti-Inflammatory Effects of Rivaroxaban vs. Dabigatran in Patients with Non-Valvular Atrial Fibrillation (RIVAL-AF Study): Multicenter Randomized Study. Heart Vessel. 2019, 34, 1002–1013. [Google Scholar] [CrossRef]
  100. Han, N.; Jin, K.; He, K.; Cao, J.; Teng, L. Protease-Activated Receptors in Cancer: A Systematic Review. Oncol. Lett. 2011, 2, 599–608. [Google Scholar] [CrossRef]
  101. DeFeo, K.; Hayes, C.; Chernick, M.; Van Ryn, J.; Gilmour, S.K. Use of Dabigatran Etexilate to Reduce Breast Cancer Progression. Cancer Biol. Ther. 2010, 10, 1001–1008. [Google Scholar] [CrossRef] [PubMed]
  102. Alexander, E.T.; Minton, A.R.; Hayes, C.S.; Goss, A.; Van Ryn, J.; Gilmour, S.K. Thrombin Inhibition and Cyclophosphamide Synergistically Block Tumor Progression and Metastasis. Cancer Biol. Ther. 2015, 16, 1802–1811. [Google Scholar] [CrossRef] [PubMed]
  103. Alexander, E.T.; Minton, A.R.; Peters, M.C.; Van Ryn, J.; Gilmour, S.K. Thrombin Inhibition and Cisplatin Block Tumor Progression in Ovarian Cancer by Alleviating the Immunosuppressive Microenvironment. Oncotarget 2016, 7, 85291–85305. [Google Scholar] [CrossRef]
  104. Vianello, F.; Sambado, L.; Goss, A.; Fabris, F.; Prandoni, P. Dabigatran Antagonizes Growth, Cell-cycle Progression, Migration, and Endothelial Tube Formation Induced by Thrombin in Breast and Glioblastoma Cell Lines. Cancer Med. 2016, 5, 2886–2898. [Google Scholar] [CrossRef]
  105. Shi, K.; Damhofer, H.; Daalhuisen, J.; Ten Brink, M.; Richel, D.J.; Spek, C.A. Dabigatran Potentiates Gemcitabine-Induced Growth Inhibition of Pancreatic Cancer in Mice. Mol. Med. 2017, 23, 13–23. [Google Scholar] [CrossRef]
  106. Yavuz, C.; Caliskan, A.; Karahan, O.; Yazici, S.; Guclu, O.; Demirtas, S.; Mavitas, B. Investigation of the Antiangiogenic Behaviors of Rivaroxaban and Low Molecular Weight Heparins. Blood Coagul. Fibrinolysis 2014, 25, 303–308. [Google Scholar] [CrossRef]
  107. Graf, C.; Wilgenbus, P.; Pagel, S.; Pott, J.; Marini, F.; Reyda, S.; Kitano, M.; Macher-Göppinger, S.; Weiler, H.; Ruf, W. Myeloid Cell–Synthesized Coagulation Factor X Dampens Antitumor Immunity. Sci. Immunol. 2019, 4, eaaw8405. [Google Scholar] [CrossRef]
  108. Guasti, L.; Squizzato, A.; Moretto, P.; Vigetti, D.; Ageno, W.; Dentali, F.; Maresca, A.M.; Campiotti, L.; Grandi, A.M.; Passi, A. In Vitro Effects of Apixaban on 5 Different Cancer Cell Lines. PLoS ONE 2017, 12, e0185035. [Google Scholar] [CrossRef] [PubMed]
  109. Kubat, E.; Onur, M.A.; Gürpınar, Ö.A. Cytotoxic Activity of Apixaban on HeLa Cells: An In Vitro Study. Hacet. J. Biol. Chem. 2018, 3, 395–402. [Google Scholar] [CrossRef]
  110. Featherby, S.; Madkhali, Y.; Maraveyas, A.; Ettelaie, C. Apixaban Suppresses the Release of TF-Positive Microvesicles and Restrains Cancer Cell Proliferation through Directly Inhibiting TF-FVIIa Activity. Thromb. Haemost. 2019, 119, 1419–1432. [Google Scholar] [CrossRef]
  111. Hiramoto, K.; Akita, N.; Nishioka, J.; Suzuki, K. Edoxaban, a Factor Xa-Specific Direct Oral Anticoagulant, Significantly Suppresses Tumor Growth in Colorectal Cancer Colon26-Inoculated BALB/c Mice. TH Open 2023, 07, e1–e13. [Google Scholar] [CrossRef]
  112. Salmi, S.; Hälinen, K.; Lin, A.; Suikkanen, S.; Jokelainen, O.; Rahunen, E.; Siiskonen, H.; Pasonen-Seppänen, S. The Association of CD8+ Cytotoxic T Cells and Granzyme B+ Lymphocytes with Immunosuppressive Factors, Tumor Stage and Prognosis in Cutaneous Melanoma. Biomedicines 2022, 10, 3209. [Google Scholar] [CrossRef]
  113. Roberts, E.W.; Broz, M.L.; Binnewies, M.; Headley, M.B.; Nelson, A.E.; Wolf, D.M.; Kaisho, T.; Bogunovic, D.; Bhardwaj, N.; Krummel, M.F. Critical Role for CD103+/CD141+ Dendritic Cells Bearing CCR7 for Tumor Antigen Trafficking and Priming of T Cell Immunity in Melanoma. Cancer Cell 2016, 30, 324–336. [Google Scholar] [CrossRef]
  114. Casu, B.; Vlodavsky, I.; Sanderson, R.D. Non-Anticoagulant Heparins and Inhibition of Cancer. Pathophysiol. Haemos. Thromb. 2007, 36, 195–203. [Google Scholar] [CrossRef] [PubMed]
  115. Borsig, L. Heparin as an Inhibitor of Cancer Progression. In Progress in Molecular Biology and Translational Science; Elsevier: Amsterdam, The Netherlands, 2010; Volume 93, pp. 335–349. ISBN 978-0-12-381282-7. [Google Scholar]
  116. Beurskens, D.M.H.; Huckriede, J.P.; Schrijver, R.; Hemker, H.C.; Reutelingsperger, C.P.; Nicolaes, G.A.F. The Anticoagulant and Nonanticoagulant Properties of Heparin. Thromb. Haemost. 2020, 120, 1371–1383. [Google Scholar] [CrossRef] [PubMed]
  117. Hostettler, N.; Naggi, A.; Torri, G.; Ishai-Michaeli, R.; Casu, B.; Vlodavsky, I.; Borsig, L. P-selectin- and Heparanase-dependent Antimetastatic Activity of Non-anticoagulant Heparins. FASEB J. 2007, 21, 3562–3572. [Google Scholar] [CrossRef] [PubMed]
  118. Ludwig, R.J.; Boehme, B.; Podda, M.; Henschler, R.; Jager, E.; Tandi, C.; Boehncke, W.-H.; Zollner, T.M.; Kaufmann, R.; Gille, J. Endothelial P-Selectin as a Target of Heparin Action in Experimental Melanoma Lung Metastasis. Cancer Res. 2004, 64, 2743–2750. [Google Scholar] [CrossRef] [PubMed]
  119. Pan, Y.; Li, X.; Duan, J.; Yuan, L.; Fan, S.; Fan, J.; Xiaokaiti, Y.; Yang, H.; Wang, Y.; Li, X. Enoxaparin Sensitizes Human Non–Small-Cell Lung Carcinomas to Gefitinib by Inhibiting DOCK1 Expression, Vimentin Phosphorylation, and Akt Activation. Mol. Pharmacol. 2015, 87, 378–390. [Google Scholar] [CrossRef] [PubMed]
  120. Pfankuchen, D.B.; Baltes, F.; Batool, T.; Li, J.-P.; Schlesinger, M.; Bendas, G. Heparin Antagonizes Cisplatin Resistance of A2780 Ovarian Cancer Cells by Affecting the Wnt Signaling Pathway. Oncotarget 2017, 8, 67553–67566. [Google Scholar] [CrossRef] [PubMed]
  121. Li, H.L.; Ye, K.H.; Zhang, H.W.; Luo, Y.R.; Ren, X.D.; Xiong, A.H.; Situ, R. Effect of Heparin on Apoptosis in Human Nasopharyngeal Carcinoma CNE2 Cells. Cell Res. 2001, 11, 311–315. [Google Scholar] [CrossRef]
  122. Shen, X.; Fang, J.; Lv, X.; Pei, Z.; Wang, Y.; Jiang, S.; Ding, K. Heparin Impairs Angiogenesis through Inhibition of MicroRNA-10b. J. Biol. Chem. 2011, 286, 26616–26627. [Google Scholar] [CrossRef]
  123. Ilan, N.; Elkin, M.; Vlodavsky, I. Regulation, Function and Clinical Significance of Heparanase in Cancer Metastasis and Angiogenesis. Int. J. Biochem. Cell Biol. 2006, 38, 2018–2039. [Google Scholar] [CrossRef]
  124. Zhang, J.; Zhang, Y.L.; Ma, K.X.; Qu, J.M. Efficacy and safety of adjunctive anticoagulation in patients with lung cancer without indication for anticoagulants: A systematic review and meta-analysis. Thorax 2013, 68, 442–450. [Google Scholar] [CrossRef] [PubMed]
  125. Falco, L.; Tessitore, V.; Ciccarelli, G.; Malvezzi, M.; D’Andrea, A.; Imbalzano, E.; Golino, P.; Russo, V. Antioxidant Properties of Oral Antithrombotic Therapies in Atherosclerotic Disease and Atrial Fibrillation. Antioxidants 2023, 12, 1185. [Google Scholar] [CrossRef] [PubMed]
  126. Russo, V.; Fabiani, D. Put out the Fire: The Pleiotropic Anti-Inflammatory Action of Non-Vitamin K Oral Anticoagulants. Pharmacol. Res. 2022, 182, 106335. [Google Scholar] [CrossRef] [PubMed]
Table 2. Anti-inflammatory effects of apixaban.
Table 2. Anti-inflammatory effects of apixaban.
AuthorProtocolTargetDisease ModelResults
Ishibashi [88]Pre-clinical in vitroHuman kidney mesangial cellsCitrated human plasma-induced ROS generation and adhesion molecule expression↓ MCP-1, ICAM-1
Nakase [90]OPS44 patients with ICS-↓ IL-6, hs CRP
Torramade-Moix [89]Pre-clinical in vitroHUVECs and HMEC-1Exposition to serum from uremic patients undergoing peritoneal dialysis↓ ICAM-1, VCAM-1
ROS: reactive oxygen species; MCP-1: monocyte chemoattractant protein-1; ICAM-1: intercellular adhesion molecule-1; OPS: observational prospective study; IL-6: Interleukin-6; hsCRP: high-sensitivity C reactive protein; ICS: ischemic cardioembolic stroke; HUVECs: human umbilical vein endothelial cells; HMEC-1: human microvascular endothelial cells; VCAM-1: vascular adhesion molecule-1.
Table 3. Anti-inflammatory effects of edoxaban.
Table 3. Anti-inflammatory effects of edoxaban.
AuthorProtocolTargetDisease ModelResults
Fang [91]Pre-clinical in vitroMale wild-type mice and HK-2 cells5/6 nephrectomy surgery↓ TNFa
↓ MCP-1
Candido [92]CC study42 DVT patients-↓ IL-6
HK-2: human kidney 2 cells; TNFa: tumor necrosis factor alpha; MCP-1: monocyte chemoattractant protein-1; CC: case control; DVT: deep-vein thrombosis; IL: Interleukin.
Table 4. Anti-inflammatory effects of dabigatran.
Table 4. Anti-inflammatory effects of dabigatran.
AuthorProtocolTargetDisease ModelResults
Tripathy [94]Pre-clinical in vitroRat brain endothelial cell culturesHypoxia exposition↓ MCP-1
Song [97]Pre-clinical in vitroMale New Zealand white rabbitsCoronary-occlusion-induced acute myocardial infarction↓ TNFa, IL-1
↓ NF-kB
Mahmoud [95]Pre-clinical in vitroAdult male albino ratsCCl4-induced liver fibrosis↓ TNFa, IL-1
Saifi [96]Pre-clinical in vitroMale Swiss albino miceUUO-induced renal fibrosis↓ TNFa, IL-1
Rahadian [98]Pre-clinical in vivoSTZ-induced diabetic C57BL/6 J miceSTZ-induced diabetes↓ MCP-1, ICAM-1
Kikuchi [99]RCT117 AF patients-↓ IL-6, IL-18
MCP-1: monocyte chemoattractant protein-1; TNFa: tumor necrosis factor alpha; CCl4: carbon tetrachloride; UUO: unilateral ureteral obstruction; STZ: streptozotocin; IL-1: Interleukin-1; NF-kB: nuclear transcription factor kB; ICAM-1: intercellular adhesion molecule-1; IL-6: Interleukin-6; IL-8: Interleukin-8; AF: atrial fibrillation; RCT: randomized controlled trial.
Table 5. Anticancer effects of DOACs.
Table 5. Anticancer effects of DOACs.
AuthorProtocolDOACTargetDisease ModelResults
DeFeo [101]Pre-clinical in vitro and in vivoDabigatranMDA-MB-231 cells, 4T1 cells and BALB/c miceBreast cancer↓ Cell migration both in vitro and in vivo
↓ Circulating 4T1 cells
↓ Tumor dimension
↓ Liver micrometastases
Alexander [102]Pre-clinical in vitro and in vivoDabigatran4T1 cells and BALB/c miceBreast cancer↓ Tumor dimension
↓ Lung metastases
↓ TGFβ
↓ Arginase + Gr-1 + CD11b+ MDSCs
↓ splenomegaly
↓ Circulating TF microparticles
↓ Activated platelets
Alexander [103]Pre-clinical in vitro and in vivoDabigatranID8-luc mouse ovarian carcinoma cells andC57/Bl6 miceOvarian cancer↓ Tumor growth
↓ Ascites ↓ Circulating activated platelets ↓ Circulating TF microparticles ↓ Gr1+/CD11b+ MDSCs
↓ CD11b+/CD11c+ DCs
↓ TGF-β, VEGF, IL-6, IL-10, and MCP-1↑ IFN-g
Vianello [104]Pre-clinical in vitroDabigatranMDA-MB-231 and U87-MGBreast cancer, glioblastoma↓ Thrombin-induced:
—Tumor proliferation
—p27 downregulation
—Cyclin D1 upregulation—Proangiogenetic protein expression
—Vascular tube formation
—Cell migration
Shi [105]Pre-clinical in vitro and in vivoDabigatranPanc02 cells and C57Bl/6 micePancreatic cancer↓ Tumor growth
↓ Cell dissemination
Yavuz [106]Pre-clinical in vivoRivaroxabanRoss 308 strain fertilized hen eggsCAM↓ Angiogenesis
Graf [107]Pre-clinical in vitro and in vivoRivaroxabanMelanoma B16F10 cells, fibro-sarcoma T241, PyMT tumor cells, and mutant miceBreast cancer, melanoma, fibrosarcoma↓ Tumor growth
↓ Metastases
↓ TAMs expression of Mrc1 CD204
↓ FoxP3 + CD4+ T cells in TME
↑ Tumor-killing granzyme B+ CD8+ T cells
↑ CD169+ macrophages and CD8+ DCs
↑ Antitumor immunity
Guasti [108]Pre-clinical in vitroApixabanOVCAR3, MDA-MB-231, CaCO-2, LNCaP, U937 cellsOvarian cancer, breast cancer, colon cancer, prostate cancer, histiocytic lymphoma↑ p16
↑ Cell mortality (OVCAR3, MDA-MB-231, CaCO-2, U937 cells)
↓ Cell migration (OVCAR3, CaCO-2)
↓ Cell Proliferation (OVCAR3, CaCO-2, LNCaP)
Kubat [109]Pre-clinical in vitroApixabanHeLa cellsCervical cancer↓ Cell viability
Featherby [110]Pre-clinical in vitroApixabanMDA-MB-231 and AsPC-1 cellsBreast cancer, pancreatic cancer↓ Release of TF-microvesicles from both fXa-activated cells and non-activated cells
↓ Cell proliferation of both fXa-activated cells and non-activated cells
Hiramoto [111]Pre-clinical in vitro and in vivoEdoxabanColon26 cells and BALB/c miceColon cancer↓ Tumor growth
↓ TF, PAI-1, IL-6, and MMP-2
↓ PAR2, STAT3, cyclin D1, and Ki67
MDSCs: myeloid-derived suppresser cells; DCs: dendritic cells; TAM: tumor-associated macrophages; TME: tumor microenvironment; CAM: chick chorioallantoic membrane; TF: tissue factor; PAI-1: plasminogen activator inhibitor 1; MMP-2: metalloproteinase 2; PAR2: protease-activated receptor 2; STAT3: signal transducer and activator of transcription 3.
Table 6. Anticancer effects of heparins.
Table 6. Anticancer effects of heparins.
AuthorProtocolTargetDisease ModelResults
Hostettler [117]Pre-clinical in vitro and in vivoMC-38, B16-BL6 cells and C57BL/6 J miceColon carcinoma/melanoma↓ Metastases
Ludwig [118]Pre-clinical in vitro and in vivoB16F10 cells and C57BL/6 J miceMelanoma↓ Metastases
Pan [119]Pre-clinical in vitro and in vivoNSCLC-A549, H1975 cells and BALB/c miceNon-small-cell lung cancer↓ Tumor volume, cell migration
Pfankuchen [120]Pre-clinical in
vitro
A2780 cellsOvarian cancer↑ Cisplatin
sensitivity
Li [121]Pre-clinical in vitroCNE2 cellsNasopharyngeal carcinoma↑ DNA fragmentation, apoptosis, bax/bcl2 ratio
Shen [122]Pre-clinical in vitroHMEC-1 cells-↓ MiR-10b expression
↓ Cell migration, tube formation, and angiogenesis
MC-38: murine cell line of colorectal cancer; B16-BL6: murine cell line of melanoma; NSCLC-A549: non-small-cell lung cancer cisplatin-resistant cell lines; H1975: human cell line of lung adenocarcinoma; A2780: human cell line of ovarian cancer; CNE2: human cell line of nasopharyngeal carcinoma; HMEC-1: human microvascular endothelial cells.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Russo, V.; Falco, L.; Tessitore, V.; Mauriello, A.; Catapano, D.; Napolitano, N.; Tariq, M.; Caturano, A.; Ciccarelli, G.; D’Andrea, A.; et al. Anti-Inflammatory and Anticancer Effects of Anticoagulant Therapy in Patients with Malignancy. Life 2023, 13, 1888. https://doi.org/10.3390/life13091888

AMA Style

Russo V, Falco L, Tessitore V, Mauriello A, Catapano D, Napolitano N, Tariq M, Caturano A, Ciccarelli G, D’Andrea A, et al. Anti-Inflammatory and Anticancer Effects of Anticoagulant Therapy in Patients with Malignancy. Life. 2023; 13(9):1888. https://doi.org/10.3390/life13091888

Chicago/Turabian Style

Russo, Vincenzo, Luigi Falco, Viviana Tessitore, Alfredo Mauriello, Dario Catapano, Nicola Napolitano, Moiz Tariq, Alfredo Caturano, Giovanni Ciccarelli, Antonello D’Andrea, and et al. 2023. "Anti-Inflammatory and Anticancer Effects of Anticoagulant Therapy in Patients with Malignancy" Life 13, no. 9: 1888. https://doi.org/10.3390/life13091888

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