You are on page 1of 19

Vol.

9 (2024) 37
Cardiovascular Innovations and Applications ISSN 2009-8618
DOI 10.15212/CVIA.2024.0013

REVIEW

Precision Monitoring of Antithrombotic Therapy


in Cardiovascular Disease
Meng Yuan1, Haichu Wen2, Yuan Wang2 and Jie Du2
1
Cardiovascular Biology Laboratory, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
2
Cardiovascular Biology Laboratory, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen ­Hospital,
Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education &
­Collaborative Innovation Center for Cardiovascular Disorder, Beijing 100029, China
Received: 16 November 2023; Revised: 20 January 2024; Accepted: 22 January 2024

Abstract
Thrombosis, the process of blood clot formation in blood vessels, is an important protective mechanism for avoiding
excessive blood spillage when an individual is exposed to trauma. The body has both a thrombosis inhibition and a
thrombus removal system, which interact in a balanced manner. If these mechanisms become unbalanced, and too
many clots form and block the lumen, thrombosis occurs. Thrombosis is currently the leading cause of death from
disease in humans and is one of the most common events leading to many cardiovascular diseases. Antithrombotic
drugs are an integral part of the pharmacological treatment regimens, and interventional strategies are currently recom-
mended for thrombotic complications in patients with thrombosis. Despite major advances in these therapies, the high
risk associated with thrombosis and bleeding remains, because of the complex interplay among patient comorbidities,
drug combinations, multifaceted dose adjustments, and care settings. Detailed assessment of the effects of bleeding
and thrombosis is necessary to establish optimal treatment plans for patients with thrombosis. This study retrospec-
tively evaluated methods for assessing the risk of bleeding/ischemia in thrombosis and the individualized use of these
methods.

Keywords: cardiovascular disease; antithrombotic therapy; individualized monitoring

Introduction movement of a thrombus within the bloodstream,


may potentially cause vessel blockage and result in
Thrombosis is a common condition that severely tissue hypoxia, necrosis, or edema. This collective
endangers human health [1]. This pathological con- process, termed thrombosis, is categorized into arte-
dition occurs when a blood clot (embolus) forms rial or venous thrombosis, according to the affected
and obstructs blood vessels, thereby impairing cir- vessel type [2, 3]. Arterial thrombosis can lead to
culation. Thromboembolism, the detachment and life-threatening diseases, such as myocardial infarc-
tion and cerebral infarction, and venous thrombosis
can lead to complications such as pulmonary embo-
lism and deep vein thrombosis. Standard anticoagu-
Correspondence: Yuan Wang and Jie Du, Beijing
Anzhen Hospital, No. 2 Anzhen Rd, Chaoyang District, lant therapy for formed thrombi is an important tool
Beijing 100029, China, E-mail: wangyuan980510@163.com; to decrease morbidity and mortality [4]. However,
jiedu@ccmu.edu.cn because of interindividual differences in metabolic

© 2024 Cardiovascular Innovations and Applications. Creative Commons Attribution-NonCommercial 4.0 International License
2 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

processes and pharmacodynamic responsiveness


to anticoagulants, even with standard treatment in Vein
accordance with guidelines, some patients experi-
ence secondary complications, such as severe gas-
trointestinal bleeding or ischemic events. Therefore, Artery
precise individualized anticoagulation therapy is
essential.
Thrombosis
Anticoagulation effects can be monitored through K - MA
R

Amplitude
LY30
the detection of indicators such as prothrombin time angle

(PT), activated partial thromboplastin time (APTT),


and international normalized ratio (INR) [5], and Clot Formation Fibrinolysis
treatment plans can be adjusted to some extent. Time
However, these indicators cannot adequately reflect
interindividual differences in the status of the coag-
ulation system. In recent years, new technologies
such as thromboelastography have brought hope for
Figure 1 Timeline for the Evaluation of Hemorrhage and
individualized treatment; these methods can detect Ischemia Detection Techniques.
multiple dimensions, such as thrombin generation R: reaction time, K: setting time, αAngle: blood clot forma-
and platelet aggregation force, thus aiding in the tion rate, MA: maximum clot intensity, LY30: dissolution
assessment of individual coagulation function [6] degree of blood clot.
and providing a basis for formulating precise anti-
coagulation treatment intensity. In addition, new individualization, and the future outlook, to promote
tools such as genetic testing have shown promise the development of this field, and the improvement of
[7] by enabling the identification of individual dif- the effectiveness and safety of anticoagulation therapy.
ferences in patients’ prothrombin activity, platelet
adhesion capability, and other relevant functions.
Some studies have also sought to establish new Classification, Epidemiology, and
scoring ­systems that include clinical factors to guide Status of Thrombosis Treatments
individualized medication.
However, truly accurate and individualized antico- According to the site of thrombus formation, throm-
agulation treatment protocols and guidelines remain bosis can be classified into two main categories:
lacking, because of the following reasons: the tests arterial thrombosis and venous thrombosis. Arterial
reflect only a portion of coagulation function, thus thrombosis occurs primarily in the cardiovascular
making the assessment incomplete; no operational system, and common forms of arterial thrombosis
scoring system incorporating clinical characteristics include coronary artery thrombosis and cerebral
is available; and consideration of age, renal function, artery thrombosis [8]. Arterial thrombosis directly
and other differences in the development of individ- blocks blood vessels and causes tissue necrosis,
ualized monitoring and medication regimens is lack- thereby leading to myocardial infarction, cerebral
ing. Therefore, new comprehensive and accurate infarction, and other life-threatening diseases in
testing technology must be developed and used in severe cases [9]. Venous thrombosis, which occurs
combination with clinical characteristics to establish primarily in the deep veins of the lower limbs and
personalized treatment and ­monitoring strategies as leads to varicose veins and skin color changes, can
illustrated by Figure 1. Only in this way can anti- also cause dangerous pulmonary embolism if the
coagulation therapy be transformed from traditional thrombus dislodges in the pulmonary artery [10].
group monitoring to truly individualized monitor- According to epidemiological data, the morbidity
ing, to benefit more patients with thrombosis. and mortality of thrombotic diseases are high. More
In this article, we review dilemmas in the field than 3 million venous thrombosis events occur per
of anticoagulation therapy for thromboembolic year in China, and more than 200,000 patients die
disease, existing progress in achieving treatment from pulmonary embolism [11]. Arterial thrombosis
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 3

is also extremely common, and is one of the leading damage when blood flow is stagnant, and exacer-
causes of stroke and myocardial infarction [12]. The bates the coagulation response [16]. In addition,
incidence of thrombosis is also high in developed coagulation products such as D-dimers directly
countries: each year, approximately 600,000 peo- stimulate endothelial cells and impair endothelial
ple in the United States experience venous throm- anticoagulation [17]. Therefore, venous thrombosis
bosis, and more than 100,000 die from pulmonary equally involves activation of the coagulation sys-
embolism [13]. Hence, thrombosis has become a tem, platelet aggregation, and vascular endothelial
major threat to human health, and in-depth research damage [18].
on the mechanism of thrombus formation must be Current clinical guidelines recommend initiat-
strengthened, to develop effective methods for pre- ing antithrombotic therapy as soon as possible after
dicting thrombus formation, and design appropriate the confirmation of thrombosis. Commonly used
preventive measures. anticoagulant or antithrombotic drugs include low-
Arterial thrombus formation is an extremely com- molecular-weight heparin, warfarin, aspirin, and
plex process that involves several key aspects, such P2Y12 inhibitors. The main mechanism of action
as damage to vascular endothelial cells, platelet of anticoagulant drugs involves inhibition of the
activation and aggregation, thrombin generation generation and activation of thrombin, and decreas-
and activation, and coagulation protein synthesis ing the amount of fibrin generated by thrombin
and deposition [9]. When vascular endothelial cells cleavage of fibrinogen, to inhibit de novo thrombus
are damaged by various factors, the tissue factor formation and dissolve the existing thrombus [19].
under the vascular endothelium is exposed and acti- The main mechanism of action of antiplatelet drugs
vates plasminogen, which in turn is converted into involves inhibition of platelet aggregation, thereby
thrombin. Thrombin cleaves fibrinogen and cata- preventing adenosine diphosphate–induced plate-
lyzes its polymerization, thereby producing fibrin, let aggregation and increasing cyclic adenosine
and promoting platelet adhesion and aggregation. monophosphate levels, and consequently inhibiting
Activated platelets also release large amounts of platelet activation and aggregation [20].
procoagulant substances, which further activate Numerous clinical studies have demonstrated
thromboplastin and accelerate thrombin production that standard anticoagulation therapy significantly
[14]. The large amount of generated thrombin can decreases patient mortality rates after thrombosis
cleave more fibrinogen and activate procoagulant and the incidence of serious thrombosis-related
factors, such as coagulation factor V and coagula- complications, such as pulmonary embolism and
tion factor VIII, which amplify the entire coagula- cerebral infarction [21, 22]. Therefore, anticoagu-
tion reaction and ultimately lead to large amounts of lation has become the standard of care for throm-
fibrin deposition, platelet aggregation, and throm- botic disorders, and guidelines recommend initiat-
bus formation [15]. Therefore, arterial thrombosis ing anticoagulation therapy immediately after the
occurs through interactions among the coagulation diagnosis of thrombosis. Although anticoagula-
system, platelets, and vascular endothelium. tion therapy is recommended by the guidelines, a
The mechanism of venous thrombosis is simi- small number of patients with thrombosis experi-
lar to that of arterial thrombosis, and involves a ence severe secondary bleeding or ischemic events
hypercoagulable state of the blood, platelet activa- [23–32]. These adverse outcomes may be associ-
tion, and endothelial damage. As the venous blood ated with the f­ollowing factors: interindividual
flow slows, blood tends to aggregate and activate differences in thrombin generation and platelet
­
the coagulation system. When the production of adhesion and aggregation, which prevent stand-
thrombin and fibrinogen increases, platelet aggre- ard-dose anticoagulation from achieving effective
gation is promoted, and a white thrombus is formed. anticoagulation; inability of some large thrombi
Simultaneously, activated platelets release procoag- to be completely dissolved, and risk of dislodg-
ulant substances that continue to activate t­hrombin ing residual thrombi; and insufficient monitoring
production and promote red blood cell thrombus of coagulation recovery during anticoagulation
formation. The poor elasticity of the venous wall therapy, thus precluding timely adjustment of the
makes vascular endothelial cells susceptible to therapeutic regimen (Table 1).
4

Table 1 Randomized Clinical Trials on Antiplatelet Therapy in Patients with Thrombotic Diseases.

Study Size Follow up Treatment Primary endpoint Outcome


ARAMIS [23] 163,038 36 hours NOAC Intracranial hemorrhage 3.7% vs 3.2%;
RR −0.51 (−1.36 to 0.34)
NCDR LAAO [24] 31,994 45 days and Aspirin vs warfarin vs Incidence of major adverse events, any 36.9% vs 20.8% vs
6 months DOAC vs DAPT stroke or transient ischemic attack, and 13.5% vs 12.3%;
readmission events HR 0.692 (0.569–0.841)
ACTIV-4B [25] 657 45 days Aspirin vs apixaban All causes of death, symptomatic venous, 2.0% vs 4.5%;
arterial thromboembolism, stroke, or MI RR 0.2 (−0.27 to 0.68)
INSPIRATION [26] 562 30 days Intermediate-dose Composite of venous or arterial 45.7% vs 44.1%;
enoxaparin vs standard thrombosis, treatment with OR 1.0 (0.76–1.48)
enoxaparin extracorporeal membrane oxygenation,
or mortality within 30 days
Voyager PAD [27] 6564 3 years Aspirin vs levofloxacin First occurrence of acute lower limb HR 0.86 (0.75–0.98)
+ aspirin ischemia, vascular major amputation, MI,
ischemic stroke, or cardiovascular death
MARINER [28] 4909 45 days Rivaroxaban vs Symptomatic VTE, non-hemorrhagic 1.28% vs 1.77%;
placebo stroke, cardiovascular death, or MI RR 0.8 (0.04–0.91)
TO-ACT [29] 67 1 year Heparin Ratio of mRS 0–1 points RR 0.99 (0.71–1.38)
Rivaroxaban with or without aspirin 7470 21 months Oral rivaroxaban + Cardiovascular death, MI, or stroke 5% vs 7%;
in patients with stable peripheral or aspirin vs aspirin HR 0.72 (0.57–0.90)
carotid artery disease [30]
ISAR-TRIPLE [31] 614 9 months 6 weeks clopidogrel vs Composite of death, MI, definite stent 9.8% vs 8.8%;
6 months clopidogrel thrombosis, stroke, or TIMI major bleeding HR 1.14 (0.68–1.91)
PLATO [32] 18,624 1 year Ticagrelor + aspirin vs Composite of vascular death, MI, or 9.8% vs 11.7%
Clopidogrel + aspirin stroke HR (0.77–0.92)
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

CV, cardiovascular; DAPT, dual antiplatelet therapy; HR, hazard ratio; MACE, major adverse cardiovascular events; MI, myocardial infarction; OR, odds ratio; RR, rela-
tive risk; ST, stent thrombosis; TIMI, thrombolysis in myocardial infarction.
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 5

Mechanisms Underlying Bleeding/ of embolic lesions and coagulation function while


Ischemic Events performing anticoagulation therapy is important to
guide the adjustment of anticoagulation therapy and
Genetic factors are important risk factors for throm- decrease the risk of secondary adverse events.
bosis. Several inherited coagulation factor defects or For different types of thrombosis, individualized
anticoagulation factor defects, such as defects in anti- monitoring strategies are required, depending on
coagulant proteins C and S, the presence of antico- the characteristics of the condition, to prevent sec-
agulant protein antibodies, and mutations at the fac- ondary bleeding and ischemic events to the greatest
tor V locus [33, 34], increase thrombosis risk. These extent possible. Moreover, for myocardial i­ nfarction
genetic defects lead to dysfunction of the coagulation or cerebral infarction caused by acute arterial throm-
system and promote excessive coagulation. bosis, given the rapid thrombus formation and the
Infections, another common thrombotic trigger, high risk of bleeding transformation, the intensity of
can cause inflammatory responses, cytokine release, anticoagulation must be monitored to avoid exces-
activation of the coagulation system and platelets, sive anticoagulation, and prevent thrombus rupture
and damage to the vascular endothelium. Severe and bleeding. Simultaneously, cardiac enzymes,
infections can also cause dehydration and impaired electroencephalograms, and other indicators should
venous reflux, and further increase thrombosis risk be monitored, and timely adjustment of treatment
[27]. Moreover, certain pathogens directly affect plans is necessary if ischemia worsens. For deep
the coagulation system and induce thrombosis [35]. vein thrombosis of the lower extremities, residual
Novel coronaviruses directly invade the vascular thrombus size changes and D-dimer levels should
endothelium and activate platelets and coagulation be monitored to determine the effects of throm-
factors, thereby leading to virus-associated thrombo- bolysis, to avoid large residual thrombus dislodge-
sis. In addition, viral infections can initiate the body’s ment and subsequent pulmonary artery embolism.
immune-mediated coagulation response and even the Simultaneously, recovery of coagulation function
development of antibodies against anticoagulant pro- should be monitored, and the intensity of anticoagu-
teins, thus resulting in immune-associated thrombosis lation should be individualized accordingly.
[36]. These findings provide new perspectives regard- For postoperative venous thrombosis, in ­addition
ing the mechanism of viral infection–associated to standard anticoagulation therapy, monitoring
thrombosis. In recent years, new mechanisms through of the mobility of the affected limbs, promoting
which viral infections regulate the balance of the ­physical activity, and avoiding prolonged bed rest
coagulation system have received extensive research are ­necessary. For patients with underlying hyper-
attention. In infections caused by novel coronaviruses, coagulable states, long-term monitoring is needed
viral proteins activate platelets and endothelial cells, to prevent new thrombus formation after anticoagu-
and lead to abnormal release of cytokines, which in lation is discontinued. Personalized monitoring of
turn activate thrombin generation [37]. In addition, different types of thrombotic lesions can effectively
novel coronaviruses can directly induce local throm- predict and prevent secondary serious adverse
bosis by destroying the endothelial cells of pulmonary events, and guide the adjustment of anticoagulant
microvessels [38]. Potential mechanisms underlying therapy, to ensure therapeutic efficacy.
the occurrence of hemorrhagic events include exces-
sive inhibition of the coagulation system by antico-
agulant drugs, which disrupt the normal connection Updated Approaches for Predicting
between the thrombus and the vessel wall, and con- Thrombosis/Bleeding-Related
sequently lead to dislodgement of the thrombus and
Events in Venous Thromboembolism
hemorrhage. Ischemic events are associated with
mechanisms such as dislodgement of residual throm-
Definition of the Disease, Epidemiology,
bus and vessel embolization, as well as insufficient
and Guideline-Recommended Treatment
recovery of platelet function after discontinuation of
anticoagulant drugs, thus leading to re-aggregation Deep venous thrombosis is a venous return disorder
of emboli [39]. Therefore, monitoring the recovery characterized by the formation of abnormal blood
6 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

clots in the deep veins, typically in the lower extrem- and closely monitoring coagulation parameters are
ities. Dislodgement of these thrombi can lead to critical to avoid excessive anticoagulation and sub-
pulmonary embolism (PE). Together, deep venous sequent bleeding. For patients with lower bleeding
thrombosis and pulmonary embolism are referred risk, standard dosing can be applied, with continu-
to as venous thromboembolism (VTE) [40]. VTE ous monitoring to balance the risk of ischemia and
manifests at a rate of 1–2 incidents per 1000 per- serious bleeding [47]. This approach ensures a more
son-years, and has a higher prevalence in men than personalized, safer anticoagulation strategy.
women. Notably, its incidence escalates to 1 event
per 100 person-years in individuals over 55 years
Limitations of Existing Methods and the
of age [41]. The prevalence of VTE is lower in Asia
Dilemma of Detection
than in Europe and the United States. Alarmingly,
approximately 20% of patients with VTE die within Traditional prothrombin time assays indicate
1 year of diagnosis. Current guidelines advocate thrombin generation, and the APTT evaluates both
the use of novel anticoagulants, such as direct oral endogenous and exogenous thrombin activation.
anticoagulants (DOACs), which inhibit thrombin However, neither assay directly measures changes
generation and activation, thereby decreasing fibrin in fibrin levels. In contrast, iProspect technology
formation [42]. directly quantifies fibrin D-dimer levels, thus ena-
bling more dynamic assessment of fibrin ­production
Subsequent Bleeding/Ischemic Events and lysis [48]. Accurately determining anticoagula-
after Antithrombotic Therapy and Risk tion intensity and overall coagulation status requires
Stratification Tools for Patients with VTE more than a single index; instead, multiple test
results must be integrated with the clinical picture.
Olaf and colleagues have demonstrated that the inci- Current models, such as the Padua prediction in
dence of pulmonary embolism can be significantly ACCP guidelines, use primarily prothrombin time
decreased, from 20% with no treatment to 3% with and D-dimer, but fall short in incorporating crucial
standard anticoagulation therapy. Hisada et al. [43] individual factors such as age and liver or renal
have reported post-anticoagulation complications, function, which are essential for assessing bleeding
such as severe gastrointestinal bleeding, in 1.5% risk [49].
of patients. Despite standard anticoagulation treat- Recommendations for normal values of existing
ment, a small proportion of patients experience monitoring indicators are too general, and signifi-
secondary adverse events [44], such as ischemic cant differences exist among types and stages of
incidents or gastrointestinal bleeding. Therefore, venous thrombosis; however, refined and clearly
personalized risk assessment and tailored anticoag- defined guideline thresholds are lacking [33]. The
ulation strategies are needed to minimize the likeli- current limitations in the clinical applicability of
hood of such adverse events. monitoring results are evident, particularly because
The risk of adverse events after venous throm- most indicators detect primarily overt hemorrhage
bosis is influenced by patient-specific risk factors, without predicting concealed chronic blood loss.
such as age over 70 years, hepatic and renal insuf- Enhancing clinical utility requires establishing
ficiency, hypoproteinemia, and a history of acid- clearer and more stringent risk value intervals tai-
suppressing drug use. Older patients and those with lored to different disease types and stages. This
renal insufficiency face a particularly elevated risk approach would enable the conversion of monitor-
of gastrointestinal bleeding, because of diminished ing results into clinically actionable recommenda-
clearance of anticoagulants [45, 46]. Concurrent tions for adjusting treatment protocols – a crucial
conditions such as ulcerative colitis further increase step toward achieving personalized anticoagula-
this risk. Therefore, anticoagulation therapy must tion therapy. In patients with high-risk venous
be individualized according to a comprehen- thrombosis with multiple comorbidities, such as
sive risk assessment in high-risk patients, such as heart failure or malignant tumors, the coagula-
older people, or those with hepatic or renal insuf- tion status is notably complex and variable. The
ficiency. Using decreased doses of anticoagulants evident dysfunction in the blood circulation and
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 7

coagulation systems significantly affects the effi- elevated cardiac enzymes [51]. Chronic coronary
cacy and outcomes of anticoagulation therapy syndrome (CCS) refers to chronic stenotic lesions
[50]. However, the current monitoring system of the coronary arteries that result in myocardial
does not consider these key influencing factors in ischemia and hypoxia, which manifest as stable
developing targeted testing protocols and medi- angina [52]. The incidence rates of ACS and CCS
cation strategy recommendations. Therefore, the are high; notably, the incidence of CCS is two
establishment of a new comprehensive assessment times higher than that of myocardial infarction,
model and the development of novel testing indi- and CCS is projected to affect 18% of the adult
cators, to optimize individualized monitoring and population by the year 2030 [53]. Percutaneous
treatment regimen selection for patients with such coronary intervention (PCI) is recommended for
complex venous thrombosis, will be an important ACS to restore perfusion; PCI or coronary artery
direction of development for increasing the preci- bypass graft surgery is recommended for CCS, to
sion of anticoagulation therapy. increase blood flow according to the severity of
Current monitoring models have notable limita- the lesion; and antithrombotic therapy with anti-
tions in facilitating individualized anticoagulation platelet and anticoagulant drugs is recommended
therapy, and often cannot support precise assess- to prevent de novo thrombosis [54].
ment and interpretation of the coagulation status
and anticoagulation response in specific patient Bleeding/Ischemic Events after
groups. A clear need exists for a more scientific Antithrombotic Therapy and Risk
and rational scoring system incorporating patients’ Stratification Tools for Patients with ACS
individual characteristics alongside existing test- and CCS
ing indices. Moreover, therapeutic objectives and
risk criteria tailored to different types of venous According to the expert consensus report published
thrombosis and stages of disease progression must by the American Heart Association, approximately
crucially be defined. This approach would enable 5% of patients with ACS who have undergone
the development of truly personalized anticoagu- dual antithrombotic therapy with anticoagulation
lation treatment plans, thus enhancing the effec- and antiplatelets experience various degrees of
tiveness and safety of medications for patients hemorrhagic adverse events, including severe
­
with venous thrombosis. Achieving this goal will ­gastro­intestinal bleeding and cerebral hemorrhage;
necessitate close collaboration between clinicians moreover, approximately 2% of these patients expe-
and laboratory personnel, and fostering a patient- rience new ischemic cardiovascular events, such as
centered approach in the treatment of venous myocardial infarction extension and angina aggra-
thrombosis. vation [55]. In contrast, patients with CCS who
have received the same treatment have a 3% inci-
dence of bleeding events and a 7% incidence of new
Updated Approaches for Predicting ischemic events [55]. The difference in the inci-
Thrombosis/Bleeding-Related dence of secondary adverse events after treatment is
Events in Acute Coronary Syndrome associated with the pathophysiological characteris-
and Chronic Coronary Syndrome tics of both ACS and CCS [56]. In addition to the
traditional CRUSADE, Syntax, and GRACE scores
[57], the PRECISE DAPT score and the PRAISE
Definition of the Disease, Epidemiology,
score are useful in the assessment of adverse clini-
and Guideline-Recommended Treatment
cal events after hemodialysis in patients with ACS
Acute coronary syndrome (ACS) is a group of [58]. The efficacy of anticoagulants can also be
clinical syndromes in which myocardial ischemia determined by measuring plasma concentrations of
and hypoxia are caused by decreased or inter- anticoagulant drugs; e.g., the efficacy of ticagrelor
rupted coronary blood flow due to coronary can be assessed by measuring the plasma concentra-
thrombosis. Its main manifestations include tion of its main active metabolite (AR-C124910XX)
acute angina pectoris, ST-segment changes, and [59]. Traditional platelet function assays, such as
8 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

VerifyNow, light transmission aggregometry, multi- function. VASP assessment involves measuring its
electrode platelet aggregometry (MEA), and vaso- phosphorylation levels in activated platelets, which
dilator-stimulated phosphoprotein (VASP), have indicates platelet inhibition and is particularly valu-
been used to assess adverse ­ clinical events in able in evaluating the effects of antiplatelet drugs,
patients with ACS [60]. Introduced in 2005, such as P2Y12 antagonists. VASP has become a
VerifyNow is a mass s­pectrometry-based system critical tool in clinical practice for assessing the
that measures platelet aggregation. A blood sample efficacy of antiplatelet drugs and platelet function.
is mixed with a carrier containing a specific activa- This method aids in crafting personalized therapeu-
tor to stimulate platelet activation. The system eval- tic strategies and in risk assessment for thrombosis-
uates the efficacy of antiplatelet drugs by monitor- related disorders, thus advancing patient-specific
ing changes in optical signals after platelet medical care [66, 67]. The limitations of VASP,
aggregation, thereby providing a precise method for such as differences in technical standardization,
assessing the effects of antiplatelet therapy [61]. complexity, and uncertainty in the interpretation of
This method is used in clinical applications to help the results, have limited its widespread use as an
assess patients’ responses to medications, guide indicator of platelet function [68, 69]. Developed
treatment adjustments, prevent the risk of bleeding between the late 2000s and 2010, MEA is a key
during surgery, and assess patients’ risk of thrombo- method for assessing platelet function and the
sis, thus providing rapid and accurate results. effects of antiplatelet therapy. MEA replicates the
Although the VerifyNow assay has emerged as a platelet aggregation process after vascular injury
novel test of platelet function assessing platelet and uses multiple electrodes to monitor platelet
aggregation capability, its relevance to ischemic aggregation dynamics simultaneously. This tech-
events in patients with cardiovascular disease has nique is crucial in evaluating the effectiveness of
not been validated in several clinical trials con- antiplatelet drugs, predicting surgical risk, diagnos-
ducted in recent years [62–64]. First introduced in ing platelet dysfunction, and preventing cardiovas-
the early 1970s, light transmission aggregometry cular and cerebrovascular diseases in clinical
remains a frequently used method for assessing ­settings. MEA’s comprehensive approach enhances
platelet function. On the basis of the Lorentz- the understanding of platelet behavior; therefore,
Boltzmann law, optical turbidimetry is used to eval- this method is integral to modern thrombosis-­
uate platelet function by measuring the extent of associated medical practice [70–72]. Thromboe­
platelet aggregation in a solution. This method acti- lastography (TEG), an advanced technology for
vates platelets and quantifies aggregation by meas- dynamically monitoring coagulation function, sim-
uring changes in light transmission through the ulates the in vivo clotting process and records
sample before and after activation. Optical turbi- changes in blood samples placed in a specially
dimetry is widely used in various clinical domains designed cup. TEG measures various parameters,
and is instrumental in hematology, monitoring of including clotting time, thrombus formation rate,
anticoagulant therapy, and assessing cardiovascular clot strength, stability, and thrombolysis rate. These
diseases; it also plays a major role in drug develop- parameters collectively offer insights into coagula-
ment and clinical trials. This enduring method tion and fibrinolytic functions, and provide a more
exemplifies the integration of classic principles with comprehensive assessment than traditional coagu-
modern medical practices [65], and serves as an lation tests. The results are depicted graphically or
important tool for assessing platelet function, diag- in chart form, thus enhancing the interpretability
nosing blood clots and platelet dysfunction, and and utility of the data in clinical settings. Therefore,
monitoring antiplatelet therapy. However, the TEG is a valuable tool in the detailed evaluation of
requirement for large samples, poor reproducibility, hemostatic processes [73], and can be used to com-
and long assay times can lead to a decrease in plate- prehensively and continuously assess the overall
let activity [64, 65]. VASP research began around effects of anticoagulants and antiplatelet agents on
1994, and initially focused on its role in cell signal- the coagulation system, dynamically predict bleed-
ing, but VASP gained widespread attention in the ing and ischemic risk, provide a basis for individu-
early 2000s for its utility in monitoring platelet alized drug adjustment, help physicians assess
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 9

patient coagulation status and function, and guide therapy based on the individual characteristics of
blood management during treatment or surgery patients with ACS and CCS, and has substantial
[74]. In one clinical trial, triple antiplatelet therapy clinical dissemination value.
(clopidogrel + aspirin + cilostazol) for patients in For the same type of thrombosis, individual dif-
the clopidogrel low-responder group, identified on ferences exist in the responsiveness to anticoagu-
the basis of TEG metrics, has been found to improve lant and antiplatelet drugs [78]. Patients who exhibit
the clinical prognosis of high-risk patients undergo- high platelet reactivity and anticoagulant resist-
ing elective PCI [75]. Overall, TEG can detect ance can be assessed with several specific tests to
changes in coagulation function holistically and guide individualized dosing regimens. Detection
dynamically, and may predict the risk of complica- of genotypes associated with drug metabolism,
tions of antithrombotic therapy; therefore, its such as CYP2C19, can be applied to predict the
­application prospects are promising. Promoting the effect of clopidogrel on antiplatelet aggregation
clinical application of TEG in patients with ACS [79]. Detection of polymorphisms of coagulation
and CCS may help achieve precision and individu- factor genes, such as F5, can be used to assess the
alization of antithrombotic therapy, minimize the anticoagulant responsiveness of warfarin [80]. In
occurrence of bleeding and ischemic events, and addition, the pharmacokinetics of antithrombotic
improve the efficacy of medication. In addition, drugs varies among individuals. Patients with,
new individualized assessment methods based on rather than without, single-nucleotide polymor-
genetic and metabolomic technologies have broad phisms in the coding region of CYP3A4/5 have
application prospects. The above methods for a 30%–40% lower ability to metabolize warfarin,
assessing ischaemic imbalance are summarized in and require adjustment of the anticoagulant dose
Figure 2. The detection of genes associated with the [4]. In patients with combined hepatic and renal
metabolism of antithrombotic drugs, such as insufficiency, which directly affects metabolic
CYP2C19 and VKORC1, can be used to identify processes and the excretion rates of anticoagulant
drug metabolism function in patients and guide the and antiplatelet drugs, blood levels of plasma-drug
selection of individualized drug regimens, and can concentration must be monitored and individual-
also detect changes in vasoactive substances, such ized to mitigate the risk of excessive accumula-
as NO and cGMP, and assess the effects of antithrom- tion, thereby preventing potential side effect such
botic therapy on vascular endothelial function [76]. as bleeding disorders or toxicity [81]. Patient age
The use of metabolomic technology to detect is another factor influencing the sensitivity to
changes in metabolite composition in patients can antithrombotic drugs. Only 50% of patients older
determine the effects of the target actions of antico- than 75 years achieve the desired INR value with
agulation and antiplatelet therapy, predict bleeding standard doses of warfarin, as compared with 80%
or ischemia risk, and provide a basis for treatment of younger patients [82].
[77]. The application of these new technological Therefore, anticoagulation lower doses are
tools may help achieve truly precise antithrombotic necessary in older patients to avoid bleeding. In

TEG VASP Verifynow


Thromboelastography (TEG) is a blood clotting The VASP (platelet activation status protein) assay is VerifyNow is a rapid, point-of-care blood test for
test used to assess and monitor the entire blood a laboratory test to assess the responsiveness of assessing the efficacy and platelet function of
clotting process. platelet inhibitory therapies, particularly clopidogrel. antiplatelet drugs such as aspirin and clopidogrel.

1948 1970 1994 2000 2005

LTA MEA
Light transmission aggregometry (LTA) is a laboratory Multi-Electrode Platelet Aggregometry (MEPA) is
test that assesses platelet function by measuring the time it an advanced blood test for assessing platelet
takes for a blood sample to form a clot in response to a aggregation function and drug effects.
specific stimulus.

Figure 2 Thromboelastography Predicts the Thrombus Formation-Dissolution Process.


10 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

Table 2 Monitoring Indicators for Anticoagulant Therapy in Patients with Acute Coronary Syndrome.

Monitoring indicators Normal range Change direction Clinical significance


Prothrombin time (s) [83] 11–13 Extend Indication of increased anticoagulation strength
APTT (s) [83] 25–40 Extend Indication of inhibition of endogenous and
exogenous thrombin production
Fibrinogen (g/L) [85] 2–4 Decrease Indication of decreased fibrinogen formation
D-dimer (mg/L) [85] < 0.5 Increase Indication of old thrombolysis
Platelet count (109/L) [85] 100–300 Decrease Indication of antiplatelet drugs to take effect

a­ ddition, thrombin generation and platelet aggre- monitoring indicators have not been fully integrated
gation in pediatric and adolescent patients also with specific disease types, thereby decreasing the
differ significantly from those in adults, and clinical operability of the monitoring results [87].
require the establishment of a specific antithrom- No specific guidelines have been established for
botic regimen [83]. Finally, female patients also patients with multiple risk factors and those under-
have specific individual characteristics. In preg- going complex surgical treatments. The changes in
nant women and those who use hormonal contra- coagulation function in these patients are complex,
ceptives, the blood is in a hypercoagulable state, and the use of standard monitoring protocols does
which may affect antithrombotic therapy [84]. not fully reflect the effectiveness of their antithrom-
Therefore, individualized antithrombotic medica- botic therapy; however, no monitoring strategy
tion and monitoring should be directed to differ- has been developed specifically for this group of
ent patients through assessment of their individual high-risk patients, thus further limiting the devel-
characteristics and clinical background informa- opment of individualized therapy [88]. Monitoring
tion. Consequently, physicians must develop a protocols must also be optimized for older patients,
specific treatment strategy for each patient to patients with systemic disease comorbidities, and
achieve truly individualized and precise medica- patients with other comorbidities, because of dif-
tion. Various monitoring indicators for anticoagu- ferences in drug metabolism. Owing to differences
lant therapy in patients with ACS are compiled in in the pharmacokinetic and pharmacodynamic pro-
Table 2. cesses in these patients, the monitoring protocol
must be adjusted to guide individualized medica-
Limitations of Existing Methods and the tion. However, the applicability of the existing mon-
Dilemma of Detection itoring indexes to this patient group must be further
verified [89]. The DAPT score and the PRECISE-
Current coagulation function testing and antithrom- DAPT score rely excessively on clinical character-
botic therapy monitoring systems have several istics and do not sufficiently consider coagulation
shortcomings. First, inconsistencies exist among indexes; consequently, their value in guiding of
testing methods, and the interpretation of the results individualized medication administration is lim-
must be comprehensive. Among platelet aggrega- ited. The DAPT score includes clinical factors such
tion function tests, the VerifyNow test and VASP as age, smoking, and diabetes, and the PRECISE-
test reflect only the inhibition effect of the adenosine DAPT score includes laboratory indexes such as
diphosphate pathway and P2Y12 receptor, whereas hemoglobin and white blood cell count, but nei-
TEG can detect the alteration of platelet aggrega- ther includes coagulation function indicators such
tion function in multiple pathways. The results of as thrombin generation and platelet aggregation
these assays are not completely consistent and must force in the assessment. Thus, these scores are not
be interpreted comprehensively [86]. For each test comprehensive or sufficiently accurate in reflecting
indicator, the critical value should be set at dif- patient coagulation status. The design of individu-
ferent thresholds for different types of thrombotic alized monitoring protocols, including coagulation
diseases. However, the current risk intervals of the kinetics, platelet aggregation function, and drug
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 11

metabolism indexes of antithrombotic drugs, and Updated Approaches for


the development of actionable monitoring values Thrombosis/Bleeding-Related
in different clinical scenarios, remains topics wor- Events in Atrial Fibrillation
thy of in-depth study. A combination of basic and
clinical research will be required to establish a truly
Definition of the Disease, Epidemiology,
reliable and applicable new antithrombotic therapy
and Guideline-Recommended Treatment
monitoring system to achieve precise individual-
ized medication and management, and comparison Atrial fibrillation (AF) is a cardiac arrhythmia char-
among various methods and evaluations. acterized by rapid irregular beating of the atria [90].
In anticoagulation and antiplatelet therapy for According to epidemiological studies, in 2019,
thrombotic diseases, such as coronary artery dis- approximately 59.7 million cases of AF (including
ease, determining how to achieve accurate pre- atrial flutter) occurred worldwide, and the preva-
diction of bleeding and ischemia risks based on lence of AF in China was 2.3%. The prevalence
individual patient characteristics and conditions and incidence of AF increase gradually with age,
is key to improving the safety and effectiveness and are higher in men than women across all age
of medication. First, tools such as genetic testing groups. Most AF cases (80–90%) are nonvalvular
can be used to identify biological features such AF (NVAF), which is also the most common form
as high thrombin generation and platelet aggre- of AF in China. NVAF guidelines recommend med-
gation hyperfunction in some individuals; the ications, electrophysiological therapy, and DOACs
findings can suggest poor antithrombotic drug to decrease the risk of stroke [85, 91].
sensitivity, and enable targeting of the dosages of
anticoagulant or antiplatelet drugs. In addition,
Subsequent Bleeding/Ischemic Events
the establishment of new operable coagulation
after Antithrombotic Therapy and Risk
kinetic monitoring protocols, such as TEG test-
Stratification Tools for Patients with AF
ing, and the determination of critical values for
TEG parameters at different disease stages, can Clinical guidelines advise maintaining an INR
enable dynamic assessment of patient coagulation between 2.0 and 3.0 for patients with AF who lack
and fibrinolytic function, and guide adjustment of additional risk factors. Tailoring anticoagulation
the intensity of antithrombotic therapy. Second, or antiplatelet therapy is crucial for these patients,
individual patients’ clinical background, such as given their stroke risk. Particularly for individu-
ACS and CCS disease types, surgical modali- als with multiple stroke risk factors, such as heart
ties, and comorbidities, should be fully integrated failure, a history of hypertension, and age over 75
into the assessment of monitoring indicator find- years, guidelines strongly advocate for the use of
ings and the optimization of the antithrombotic oral vitamin K antagonists. These anticoagulants,
treatment protocols in various clinical scenarios. aimed at achieving an INR of 2.5 to 3.0 or higher,
Large-sample clinical studies are also necessary have potent anticoagulant properties and high effi-
to establish reliable bleeding and ischemia risk cacy in decreasing thrombosis risk. This targeted
assessment models to guide the selection and use approach underscores the importance of individual-
of antithrombotic drugs. ized therapy in optimizing patient outcomes in AF
In summary, only by organically combining management [91].
advanced testing technologies with extensive clini- Among patients with AF with low stroke risk,
cal experience, and developing personalized treat- antiplatelet drugs such as aspirin – which do not
ment and monitoring strategies for different patients, act directly on the coagulation system – may
can the safety and efficacy of antithrombotic therapy be considered, and their relatively low antico-
be improved while maximizing the prevention of agulant effects can decrease the risk of bleeding
bleeding and ischemic events. Cooperation among [92]. The CHA2DS2-VASc score is a guideline-­
all medical specialties will be necessary to achieve recommended tool for assessing the risk of stroke
precise individualized medication. in patients with AF. The score considers a range of
12 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

risk factors such as age, sex, hypertension, diabe- examined 1820 patients with AF combined with
tes mellitus, heart failure, stroke/transient ischemic moderate-to-severe renal insufficiency and found
attack, and vascular disease. On the basis of the that the risk of gastrointestinal hemorrhage during
total score, patients can be classified into stroke standard anticoagulation therapy was 1.7 times
risk groups, which indicate the need for anticoagu- higher than that in patients without renal impair-
lant therapy [93]. The CHA2DS2 score is similar ment [101].
to the CHA2DS2-VASc score and has also been The main indicators routinely used to moni-
shown to be useful for assessing the risk of stroke tor the effectiveness of anticoagulation therapy
in patients with AF in clinical trials [94]. The HAS- and the risk of bleeding in patients with NVAF
BLED score is used to assess the risk of bleeding in include PT, APTT, and INR. These indicators can
patients with AF during anticoagulant therapy, and be used to assess the level of thrombosis and the
considers factors such as hypertension, abnormal efficacy of anticoagulation therapy. In addition to
liver function, stroke, history of bleeding, coagula- the CHA2DS2-VASc score and the HAS-BLED
tion abnormalities, age, and drug or alcohol use. score, the R2CHADS2 score [102] adds renal
This score is used to determine whether patients are impairment and an age range of 65–74 years to
suitable for anticoagulant therapy and how best to the CHA2DS2-VASc score. The scoring system
manage the bleeding risk [95]. Routine tests such as categorizes patients with NVAF into four levels
cardiac ultrasound (echocardiography) can deter- (low, moderate, high, and very high risk), thereby
mine the presence of blood clots within the heart contributing to a more nuanced assessment of the
in patients with AF, to help assess the risk of stroke stroke risk.
and guide anticoagulant therapy [96]. Blood tests, Assessing levels of thrombosis and the efficacy of
such as INR and PT, can test coagulation function, anticoagulation therapy in patients with AF by mon-
to assess the effectiveness of anticoagulant therapy itoring coagulation pathology indexes such as PT,
and the risk of bleeding [97]. Continuous cardiac D-dimer, and APTT to guide the subsequent adjust-
monitoring, such as Holter monitoring or event ment of anticoagulation intensity can be applied to
recording, can help identify changes in the heart predict the occurrence of adverse events such as
rhythm in patients with AF and assess their risk of ischemic stroke. However, in patients with renal
bleeding/ischemia [98]. impairment leading to anticoagulant accumulation,
Because of individual differences in drug the changes in the routine coagulation indexes do
metabolism rates and sensitivity, patients with not fully reflect the precise effects of anticoagulants
NVAF have varying degrees of risk of thrombosis and therefore cannot provide reliable personalized
and gastrointestinal bleeding after treatment with medication guidance for this high-risk group; this
DOACs [99]. Special attention should be paid to aspect is one limitation of the current monitoring
patients with AF with moderate-to-severe renal system. For individuals with renal impairment, the
insufficiency. These patients are susceptible to interpretation of all coagulation indexes is diffi-
drug accumulation due to diminished glomerular cult, and actionable recommendations that can be
filtration rate, and excretion and clearance of anti- directly translated into medication and monitoring
coagulants, which may lead to severe gastrointes- protocols are lacking. These issues constrain safe
tinal hemorrhage and other adverse effects. Thus, and effective personalized anticoagulation in this
anticoagulation therapy in this group of patients is group of patients with complex AF with high stroke
highly challenging, and individualized dosing and risk and renal impairment.
monitoring measures are necessary to decrease To achieve personalized and precise anticoagu-
the risk of severe hemorrhage [100]. Many cohort lation for patients with NVAF with renal impair-
studies have reported the incidence of gastrointes- ment, while decreasing the risk of stroke and
tinal bleeding and ischemic stroke after anticoagu- controlling the risk of gastrointestinal bleeding,
lation in patients with NVAF. Lip et al. analyzed the optimal range of anticoagulant concentrations
17,633 patients with NVAF and showed that 4.9% must be defined on the basis of advanced testing
developed gastrointestinal hemorrhage, and 1.8% techniques. Moreover, establishing personalized
developed ischemic stroke [99]. Pokorney et al. medication and monitoring protocols for these
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 13

patients through prospective cohort studies, and 2.5–3.5, and minimize the probability of perivalvu-
translating them into specific clinical protocols, lar thrombosis to the greatest extent possible [105].
will be important. The optimal range of anticoagu- In contrast, a lower anticoagulation intensity regi-
lant concentrations for this group of patients can men (INR 2.0–3.0) may be chosen for patients at
be clearly defined on the basis of advanced test- low stroke risk with tissue or biologic valves [106].
ing technology. Only by combining monitoring Numerous cohort studies have reported the inci-
technology with the individual clinical character- dence of gastrointestinal bleeding and thrombotic
istics of patients and formulating individualized events due to anticoagulation therapy after heart
therapeutic strategies can the efficacy and safety valve replacement.
of anticoagulation be improved in patients with Oral anticoagulants have shown excellent anti-
NVAF combined with renal insufficiency. Close coagulant efficacy and a relatively favorable safety
collaboration among medical specialties will be profile in clinical trials and practical clinical appli-
necessary to continually optimize patient antico- cations. However, serious bleeding events occur in
agulation and monitoring. approximately 2–3.5% of patients receiving oral
anticoagulation each year [107]. These patients
may be at risk of thrombosis if anticoagulation is
Updated Approaches for Bleeding/ discontinued. Sulman et al. have reported thrombo-
Ischemic Events in Valvular Disease embolic events in 16% of patients undergoing bio-
prosthetic valve implantation and coronary artery
Definition, Prevalence, and ­Guideline- bypass grafting [107]. In patients with bioprosthetic
Recommended Treatment of Valvular valves, the risk of gastrointestinal bleeding due to
Disease overaccumulation of anticoagulants should not be
ignored, and a dynamic balance between bleeding
Valvular disease is a heart disease involving a risk and thrombosis risk must be ensured through
structural or functional abnormality of the heart regular monitoring.
valves. According to the EURO II Survey results, Currently, for the monitoring of anticoagulation
the prevalence of valve disease is 2.5% in the over- therapy in patients after heart valve replacement,
all population and increases significantly with age; conventional indicators such as PT and INR are
in the population over 75 years of age, the inci- primarily used [108]. The cardiac renal hepatic
dence rate for all valves combined reaches nearly (CRH) score, a new score combining multiple
14% [103]. The most common valve diseases are biomarkers of cardiac, renal, and hepatic func-
mitral valve closure insufficiency and aortic steno- tion (NT-proBNP, creatinine, and albumin, respec-
sis [104], and the treatment options include phar- tively), based on data from the CHINA-VHD study,
macological management, surgical repair, or valve has been identified as the key predictor of mortal-
replacement. ity in patients with valvular heart disease [109].
Cardiac ultrasound, which indicates valve activity,
Subsequent Bleeding/Ischemic Events ventricular function, chamber size, and possible
after Antithrombotic Therapy and Risk complications, can be used to assess cardiac func-
Stratification Tools for Patients with Valve tion and valve function after valve replacement.
Disease The overall pumping function of the heart can also
be assessed by measuring cardiac function indexes
Patients undergoing heart valve replacement such as the cardiac index and left ventricular ejec-
surgery require long-term oral anticoagulation
­ tion fraction [110].
therapy. Current clinical guidelines generally rec- Among mitral valve diseases, mitral valve clo-
ommend that patients undergoing mechanical sure insufficiency is the most common condition,
valve replacement surgery start taking vitamin K and TMVR is a common interventional treatment
antagonist anticoagulants such as warfarin orally in for mitral valve closure insufficiency. However,
the immediate postoperative period, to maintain a complications such as thrombosis and hemorrhage
high anticoagulation intensity with an INR value of persist in the postoperative period. The incidence
14 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

of stroke within 30 days of surgery ranges from specifically formulated monitoring strategies and
0.7% to 2.6%, and hemorrhagic complications medication recommendations are lacking for high-
occur in approximately 13% of cases [110]. risk groups, such as individuals with impaired
Among aortic valve diseases, aortic stenosis is liver and kidney function or older individuals.
the most common. In a Danish study, ischemic These issues limit the precision of anticoagulation
stroke has been found in 8% of 873,373 patients in specific patients.
[111]. Patients with mitral valve insufficiency and Long-term anticoagulation is essential for
aortic stenosis often have a combination of bleed- patients undergoing heart valve surgery but must
ing risk factors, such as congestive heart failure, be balanced against thrombosis and bleeding risk,
hypertension, advanced age, renal insufficiency, to ensure the maximum efficacy and safety of the
and diabetes mellitus. The ESC/EACTS 2017 therapy. Therefore, advanced testing techniques
and the ACC/American Heart Association 2020 and clinical experience must be combined to clearly
guidelines do not provide a clear antithrombotic define the optimal range of anticoagulant strengths
protocol, and current policies are largely depend- for different patient groups, particularly those at
ent on clinician experience. Therefore, an urgent high risk. Individualized treatment and monitoring
need exists for effective protocols to individu- plans must be developed to ensure that each patient
alize patient treatment. In a study published by attains maximum therapeutic benefit. To achieve
the JACC, a simple and practical risk score was this goal, all relevant areas of medicine must work
developed through the COAPT research database, closely together to provide precise and individual-
which consisted of four clinical variables (New ized anticoagulation for patients with heart valve
York Heart Association functional class, chronic disease.
obstructive pulmonary disease, AF or atrial flut-
ter, and chronic kidney disease) and four echo-
cardiographic variables (left ventricular ejection Conclusion and Outlook
fraction, left ventricular end-systolic internal
diameter, right ventricular systolic pressure, and This review summarized current dilemmas in
tricuspid regurgitation). The COAPT risk score antithrombotic therapy, as well as strategies for
is useful in predicting the risk of death or hospi- individualizing antithrombotic therapy by compar-
talization for heart failure in patients with severe ing risk stratification tools for various thrombotic
mitral regurgitation [112]. Elevated NT-proBNP diseases.
has been associated with elevated mortality in The primary challenge in current antithrom-
patients with moderate aortic stenosis, but no stud- botic therapy is balancing the risks of bleeding
ies have demonstrated whether early intervention and ischemia. Individual variability and uncertain
affects prognosis. In a population of patients with drug response hinder the ability to achieve con-
moderate or severe aortic stenosis, high-sensitiv- sistent and balanced therapeutic effects. Recently,
ity troponin I has been associated with increased TEG has emerged as a promising solution offer-
left ventricular mass and fibrosis on CMR imag- ing insights into thrombin generation and platelet
ing, and with aortic valve replacement and death aggregation force. This method enables accurate
[112]. Echocardiography can be used to assess the assessment of anticoagulation intensity and paves
extent of valvular disease in patients with mitral the way to precision anticoagulation therapy. In
valve closure insufficiency or aortic stenosis, to the future, TEG, particularly when integrated
assess prognosis [113]. with individual patient characteristics, is poised to
Although interventional and antithrombotic become an essential tool in monitoring anticoagu-
therapy in patients with valvular disease can effec- lation therapy.
tively improve prognosis, it cannot quantitatively In the future, with the development and in-depth
guide individualized medication use, because of application of high-throughput detection tech-
the lack of a clear definition of the optimal range nology and information technology, antithrom-
of anticoagulant intensity or blood concentra- botic therapy monitoring systems could lead to
tion targets for different populations. In addition, a shift from traditional group monitoring to truly
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 15

individualized monitoring. The high throughput Sources of Funding


and high precision of various testing technologies
may provide the best and safest precise antithrom- This work was supported by the Beijing Municipal
botic treatment plans tailored to different patients. Medical Research Institutes Pilot Reform Project
Hence, all relevant medical specialties must unite (grant number 2021-07).
in joint efforts to further improve the diagnosis
and treatment of thromboembolic diseases, as Conflict of interest
well as the efficacy and safety of antithrombotic
therapy. The authors declare no conflicts of interest.

References

1. Raskob GE, Angchaisuksiri P, 9. Lippi G, Franchini M, Targher G. by the ESC Working Group on
Blanco AN, Büller H, Gallus A, Arterial thrombus formation in Pulmonary Circulation and Right
Hunt BJ, et al. Thrombosis: a cardiovascular disease. Nat Rev Ventricular Function. COVID-19
major contributor to global disease Cardiol 2011;8(9):502–12. and thrombotic or thromboem-
burden. Semin Thromb Hemost 10. Weitz JI, Fredenburgh JC, bolic disease: implications for pre-
2014;40(7):724–35. Eikelboom JW. A test in ­ context: vention, antithrombotic therapy,
2. Alkarithi G, Duval C, Shi Y, Macrae D-dimer. J Am Coll Cardiol and follow-up: JACC state-of-
FL, Ariëns RAS. Thrombus struc- 2017;70(19):2411–20. the-art review. J Am Coll Cardiol
tural composition in Cardiovascular 11. Andreoli L, Bertsias GK, Agmon- 2020;75(23):2950–73.
Disease. Arterioscler Thromb Vasc Levin N, Brown S, Cervera R, 15. Pieters M, Wolberg AS. Fibrinogen
Biol 2021;41(9):2370–83. Costedoat-Chalumeau N, et al. and fibrin: an illustrated review.
3. Joundi RA, Menon BK. Thrombus EULAR recommendations for wom- Res Pract Thromb Haemost
composition, imaging, and outcome en’s health and the management of 2019;3(2):161–72.
prediction in acute ischemic stroke. family planning, assisted reproduc- 16. Previtali E, Bucciarelli P, Passamonti
Neurology 2021;97(20, Suppl 2): tion, pregnancy and menopause in SM, Martinelli I. Risk factors for
S68–78. patients with systemic lupus ery- venous and arterial thrombosis.
4. Mackman N, Bergmeier W, Stouffer thematosus and/or antiphospho- Blood Transfus 2011;9(2):120–38.
GA, Weitz JI. Therapeutic strate- lipid syndrome. Ann Rheum Dis 17. Dare S, Mackay DF, Pell JP.
gies for thrombosis: new targets and 2017;76(3):476–85. Relationship between smoking and
approaches. Nat Rev Drug Discov 12. Farge D, Frere C, Connors JM, obesity: a cross-sectional study of
2020;19(5):333–52. Khorana AA, Kakkar A, Ay C, 499,504 middle-aged adults in the
5. Dunois C. Laboratory monitoring of et al. International Initiative on UK general population. PLoS One
direct oral anticoagulants (DOACs). Thrombosis and Cancer (ITAC) 2015;10(4):e0123579.
Biomedicines 2021;9(5):445. Advisory Panel. 2022 international 18. Page MJ, Pretorius E. A cham-
6. Willis J, Carroll C, Planz V, clinical practice guidelines for the pion of host defense: a generic
Galgano SJ. Thromboelastography: treatment and prophylaxis of venous large-scale cause for platelet dys-
a review for radiologists and impli- thromboembolism in patients function and depletion in infec-
cations on periprocedural bleed- with cancer, including patients tion. Semin Thromb Hemost
ing risk. Abdom Radiol (NY) with COVID-19. Lancet Oncol 2020;46(3):302–19.
2022;47(8):2697–703. 2022;23(7):e334–47. 19. Sun HJ, Wu ZY, Nie XW, Bian
7. McGuire AL, Gabriel S, Tishkoff 13. O’Leary JG, Greenberg CS, Patton JS. Role of endothelial dysfunc-
SA, Wonkam A, Chakravarti A, HM, Caldwell SH. AGA clini- tion in cardiovascular diseases:
Furlong EEM, et al. The road ahead cal practice update: coagulation the link between inflammation and
in genetics and genomics. Nat Rev in cirrhosis. Gastroenterology hydrogen sulfide. Front Pharmacol
Genet 2020;21(10):581–96. 2019;157(1):34–43.e1. 2020;10:1568.
8. Chang JC. Novel classification 14. Bikdeli B, Madhavan MV, Jimenez 20. Schofield Z, Baksamawi HA,
of thrombotic disorders based on D, Chuich T, Dreyfus I, Driggin Campos J, Alexiadis A, Nash GB,
molecular hemostasis and thrombo- E, et al.; Global COVID-19 Brill A, et al. The role of valve
genesis producing primary and sec- Thrombosis Collaborative Group, stiffness in the insurgence of deep
ondary phenotypes of thrombosis. Endorsed by the ISTH, NATF, vein thrombosis. Commun Mater
Biomedicines 2022;10(11):2706. ESVM, and the IUA, Supported 2020;1(1):65.
16 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

21. Xue L, Tao L, Li X, Wang Y, Wang et al. Extended venous thrombo- 36. Shufelt C, LeVee A. Hormonal
B, Zhang Y, et al. Plasma fibrino- embolism prophylaxis in medically contraception in women with
gen, D-dimer, and fibrin degradation Ill patients: an NATF anticoagula- hypertension. J Am Med Assoc
product as biomarkers of rheumatoid tion action initiative. Am J Med 2020;324(14):1451–52.
arthritis. Sci Rep 2021;11(1):16903. 2020;133 Suppl 1:1–27. 37. Vano YA, Elaidi R, Bennamoun M,
22. Stark K, Massberg S. Interplay 29. Coutinho JM, Zuurbier SM, Chevreau C, Borchiellini D, Pannier
between inflammation and thrombo- Bousser MG, Ji X, Canhão P, Roos D, et al. Nivolumab, nivolumab-
sis in cardiovascular pathology. Nat YB, et al.; TO-ACT Investigators. ipilimumab, and VEGFR-tyrosine
Rev Cardiol 2021;18(9):666–82. Effect of endovascular treatment kinase inhibitors as first-line treat-
23. Kam W, Holmes DN, Hernandez with medical management vs stand- ment for metastatic clear-cell renal
AF, Saver JL, Fonarow GC, Smith ard care on severe cerebral venous cell carcinoma (BIONIKK): a
EE, et al. Association of recent thrombosis: the TO-ACT rand- biomarker-driven, open-label, non-
use of non-vitamin K antagonist omized clinical trial. JAMA Neurol comparative, randomised, phase
oral anticoagulants with intracra- 2020;77(8):966–73. 2 trial. Lancet Oncol 2022;23(5):
nial hemorrhage among patients 30. Anand SS, Bosch J, Eikelboom JW, 612–24.
with acute ischemic stroke treated Connolly SJ, Diaz R, Widimsky 38. Capodanno D, Mehran R,
with alteplase. J Am Med Assoc P, et al.; COMPASS Investigators. Krucoff MW, Baber U, Bhatt DL,
2022;327(8):760–71. Rivaroxaban with or without aspirin Capranzano P, et al. Defining
24. Freeman JV, Higgins AY, Wang Y, in patients with stable peripheral or strategies of modulation of anti-
Du C, Friedman DJ, Daimee UA, carotid artery disease: an interna- platelet therapy in patients with
et al. Antithrombotic therapy after tional, randomised, double-blind, coronary artery disease: a consen-
left atrial appendage occlusion in placebo-controlled trial. Lancet sus document from the Academic
patients with atrial fibrillation. J Am 2018;391(10117):219–29. Research Consortium. Circulation
Coll Cardiol 2022;79(18):1785–98. 31. Fiedler KA, Maeng M, Mehilli J, 2023;147(25):1933–44.
25. Connors JM, Brooks MM, Sciurba Schulz-Schüpke S, Byrne RA, Sibbing 39. Molander V, Bower H, Frisell
FC, Krishnan JA, Bledsoe JR, D, et al. Duration of triple therapy in T, Delcoigne B, Di Giuseppe D,
Kindzelski A, et al. ACTIV-4B patients requiring oral anticoagulation Askling J; ARTIS study group.
Investigators. Effect of antithrombotic after drug-eluting stent implantation: Venous thromboembolism with
therapy on clinical outcomes in out- the ISAR-TRIPLE trial. J Am Coll JAK inhibitors and other immune-­
patients with clinically stable symp- Cardiol 2015;65(16):1619–29. modulatory drugs: a Swedish com-
tomatic COVID-19: the ACTIV-4B 32. Wallentin L, Becker RC, Budaj A, parative safety study among patients
randomized clinical trial. J Am Med Cannon CP, Emanuelsson H, Held with rheumatoid arthritis. Ann
Assoc 2021;326(17):1703–12. C, et al. Ticagrelor versus clopi- Rheum Dis 2023;82(2):189–97.
26. Sadeghipour P, Talasaz AH, dogrel in patients with acute coro- 40. Bertsias GK, Ioannidis JP, Aringer
Rashidi F, Sharif-Kashani B, nary syndromes. N Engl J Med M, Bollen E, Bombardieri S, Bruce
Beigmohammadi MT, Farrokhpour 2009;361(11):1045–57. IN, et al. EULAR recommendations
M, et al. Effect of intermediate- 33. Valerio L, Mavromanoli AC, Barco for the management of systemic
dose vs standard-dose prophylac- S, Abele C, Becker D, Bruch L, lupus erythematosus with neuropsy-
tic anticoagulation on thrombotic et al. Chronic thromboembolic pul- chiatric manifestations: report of a
events, extracorporeal membrane monary hypertension and impair- task force of the EULAR standing
oxygenation treatment, or mortal- ment after pulmonary embolism: committee for clinical affairs. Ann
ity among patients with COVID-19 the FOCUS study. Eur Heart J Rheum Dis 2010;69(12):2074–82.
admitted to the intensive care unit: 2022;43(36):3387–98. 41. Naess IA, Christiansen SC,
the INSPIRATION randomized 34. Jovin TG, Nogueira RG, Lansberg Romundstad P, Cannegieter SC,
clinical trial. J Am Med Assoc MG, Demchuk AM, Martins SO, Rosendaal FR, Hammerstrøm J.
2021;325(16):1620–30. Mocco J, et al. Thrombectomy for Incidence and mortality of venous
27. Bauersachs RM, Szarek M, anterior circulation stroke beyond thrombosis: a population-based study.
Brodmann M, Gudz I, Debus ES, 6 h from time last known well J Thromb Haemost 2007;5(4):692–9.
Nehler MR, et al. VOYAGER PAD (AURORA): a systematic review and 42. Wolberg AS, Rosendaal FR, Weitz
Committees and Investigators. individual patient data meta-analysis. JI, Jaffer IH, Agnelli G, Baglin T,
Total ischemic event reduction with Lancet 2022;399(10321):249–58. et al. Venous thrombosis. Nat Rev
rivaroxaban after peripheral arterial 35. Büller HR, Prins MH, Lensin AW, Dis Primers 2015;1:15006.
revascularization in the VOYAGER Decousus H, Jacobson BF, Minar 43. Olaf M, Cooney R. Deep venous
PAD trial. J Am Coll Cardiol E, et al. Oral rivaroxaban for the thrombosis. Emerg Med Clin North
2021;78(4):317–26. treatment of symptomatic pul- Am 2017;35(4):743–70.
28. Barkoudah E, Piazza G, Hecht TEH, monary embolism. N Engl J Med 44. Hisada Y, Mackman N. Cancer-
Grant P, Deitelzweig S, Fang MC, 2012;366(14):1287–97. associated pathways and biomarkers
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 17

of venous thrombosis. Blood TM, Bischoff JM, et al. 2021 ACC/ randomized trial. J Am Med Assoc
2017;130(13):1499–506. AHA/SCAI guideline for coronary 2011;305(11):1097–105.
45. Watson HG, Baglin TP. Guidelines artery revascularization: executive 62. Aradi D, Gross L, Trenk D, Geisler
on travel-related venous thrombosis. summary: a report of the American T, Merkely B, Kiss RG, et al.
Br J Haematol 2011;152(1):31–4. College of Cardiology/American Platelet reactivity and clinical out-
46. Timp JF, Braekkan SK, Versteeg HH, Heart Association Joint Committee comes in acute coronary syndrome
Cannegieter SC. Epidemiology of on Clinical Practice Guidelines. patients treated with ­ prasugrel
cancer-associated venous thrombo- Circulation 2022;145(3):e4–17. and clopidogrel: a pre-specified
sis. Blood 2013;122(10):1712–23. 55. Heidenreich PA, Bozkurt B, Aguilar exploratory analysis from the
47. Kakkos SK, Gohel M, Baekgaard N, D, Allen LA, Byun JJ, Colvin TROPICAL-ACS trial. Eur Heart J
Bauersachs R, Bellmunt-Montoya MM, et al. 2022 AHA/ACC/HFSA 2019;40(24):1942–51.
S, Black SA, et al. Editor’s Choice - guideline for the management 63. Cayla G, Cuisset T, Silvain J,
European Society for Vascular Surgery of heart failure: executive sum- Leclercq F, Manzo-Silberman S,
(ESVS) 2021 Clinical Practice mary: a report of the American Saint-Etienne C, et al. Platelet
Guidelines on the Management of College of Cardiology/American function monitoring to adjust anti-
Venous Thrombosis. Eur J Vasc Heart Association Joint Committee platelet therapy in elderly patients
Endovasc Surg 2021;61(1):9–82. on clinical practice guidelines. stented for an acute coronary syn-
48. Tan R, Daneshmand A, Parys Circulation 2022;145(18):e876–94. drome (ANTARCTIC): an open-
S, Watanabe Y, Sieunarine K. 56. Lopes RD, Heizer G, Aronson R, label, blinded-endpoint, randomised
Splanchnic venous thrombosis: aeti- Vora AN, Massaro T, Mehran R, controlled superiority trial. Lancet
ologies and a review of the literature. et al. Antithrombotic therapy after 2016;388(10055):2015–22.
ANZ J Surg 2022;92(9):2224–8. acute coronary syndrome or PCI 64. Moreo A, Ambrosio G, De Chiara B,
49. van der Steen W, van de Graaf in atrial fibrillation. N Engl J Med Pu M, Tran T, Mauri F, et al. Influence
RA, Chalos V, Lingsma HF, van 2019;380(16):1509–24. of myocardial fibrosis on left ven-
Doormaal PJ, Coutinho JM, et al. MR 57. Cordero A, Rodriguez-Manero M, tricular diastolic function: noninva-
CLEAN-MED Investigators. Safety García-Acuña JM, López-Palop R, sive assessment by cardiac magnetic
and efficacy of aspirin, unfraction- Cid B, Carrillo P, et al. Additive resonance and echo. Circ Cardiovasc
ated heparin, both, or neither dur- value of the CRUSADE score to Imaging 2009;2(6):437–43.
ing endovascular stroke treatment the GRACE score for mortality risk 65. Khandhar AP, Ferguson RM, Simon
(MR CLEAN-MED): an open-label, prediction in patients with acute JA, Krishnan KM. Tailored magnetic
multicentre, randomised controlled coronary syndromes. Int J Cardiol nanoparticles for optimizing mag-
trial. Lancet 2022;399(10329): 2017;245:1–5. netic fluid hyperthermia. J Biomed
1059–69. 58. Costa F, van Klaveren D, James Mater Res A 2012;100(3):728–37.
50. Pastori D, Cormaci VM, Marucci S, S, Heg D, Räber L, Feres F, et al. 66. Schlegel N, Waschke J. Vasodilator-
Franchino G, Del Sole F, Capozza Derivation and validation of the stimulated phosphoprotein: crucial
A, et al. A comprehensive review predicting bleeding complications for activation of Rac1 in endothe-
of risk factors for venous throm- in patients undergoing stent implan- lial barrier maintenance. Cardiovasc
boembolism: from epidemiology tation and subsequent dual anti- Res 2010;87(1):1–3.
to pathophysiology. Int J Mol Sci platelet therapy (PRECISE-DAPT) 67. Tu K, Li J, Verma VK, Liu C,
2023;24(4):3169. score: a pooled analysis of individ- Billadeau DD, Lamprecht G, et al.
51. Kumar A, Cannon CP. Acute coro- ual-patient datasets from clinical Vasodilator-stimulated phosphopro-
nary syndromes: diagnosis and trials. Lancet 2017;389(10073): tein promotes activation of hepatic
management, part I. Mayo Clin Proc 1025–34. stellate cells by regulating Rab11-
2009;84(10):917–38. 59. Holm M, Tornvall P, Henareh L, dependent plasma membrane tar-
52. Knuuti J, Wijns W, Saraste A, Jensen U, Golster N, Alström P, geting of transforming growth
Capodanno D, Barbato E, Funck- et al. The MOVEMENT trial. J Am factor beta receptors. Hepatology
Brentano C, et al. 2019 ESC Guidelines Heart Assoc 2019;8(2):e010152. 2015;61(1):361–74.
for the diagnosis and management of 60. Paniccia R, Priora R, Liotta AA, 68. Bonello L, Paganelli F, Arpin-
chronic coronary syndromes. Eur Abbate R. Platelet function tests: Bornet M, Auquier P, Sampol J,
Heart J 2020;41(3):407–77. a comparative review. Vasc Health Dignat-George F, et al. Vasodilator-
53. Timmis A, Kazakiewicz D, Risk Manag 2015;11:133–48. stimulated phosphoprotein phos-
Townsend N, Huculeci R, Aboyans 61. Price MJ, Berger PB, Teirstein PS, phorylation analysis prior to per-
V, Vardas P. Global epidemiology of Tanguay JF, Angiolillo DJ, Spriggs cutaneous coronary ­ intervention
acute coronary syndromes. Nat Rev D, et al. Standard- vs high-dose for exclusion of postprocedural
Cardiol 2023;20(11):778–88. clopidogrel based on platelet func- major adverse cardiovascu-
54. Lawton JS, Tamis-Holland JE, tion testing after percutaneous coro- lar events. J Thromb Haemost
Bangalore S, Bates ER, Beckie nary intervention: the GRAVITAS 2007;5(8):1630–6.
18 M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease

69. Oelze M, Mollnau H, Hoffmann 77. Qiu S, Cai Y, Yao H, Lin C, Xie Y, thrombosis and antithrombotic
N, Warnholtz A, Bodenschatz M, Tang S, et al. Small molecule metab- therapy in whole blood. Sci Rep
Smolenski A, et al. Vasodilator- olites: discovery of biomarkers and 2020;10(1):5742.
stimulated phosphoprotein serine 239 therapeutic targets. Signal Transduct 87. McRae HL, Militello L, Refaai
phosphorylation as a sensitive moni- Target Ther 2023;8(1):132. MA. Updates in anticoagulation
tor of defective nitric oxide/cGMP 78. van Vlijmen EF, Wiewel-Verschueren therapy monitoring. Biomedicines
signaling and endothelial dysfunc- S, Monster TB, Meijer K. Combined 2021;9(3):262.
tion. Circ Res 2000;87(11):999–1005. oral contraceptives, thrombophilia 88. Kleindorfer DO, Towfighi A,
70. Morel-Kopp MC, Mullier F, Gkalea and the risk of venous thromboem- Chaturvedi S, Cockroft KM,
V, Bakchoul T, Minet V, Elalamy bolism: a systematic review and Gutierrez J, Lombardi-Hill D, et al.
I, et al. Heparin-induced multi- meta-analysis. J Thromb Haemost 2021 guideline for the prevention
electrode aggregometry method for 2016;14(7):1393–403. of stroke in patients with stroke and
­heparin-induced thrombocytope- 79. Hassouna HI. Thrombophilia and transient ischemic attack: a guideline
nia testing: communication from hypercoagulability. Med Princ Pract from the American Heart Association/
the SSC of the ISTH. J Thromb 2009;18(6):429–40. American Stroke Association. Stroke
Haemost 2016;14(12):2548–52. 80. Coen Herak D, Lenicek Krleza 2021;52(7):e364–467.
71. Tóth O, Calatzis A, Penz S, J, Radic Antolic M, Horvat I, 89. Peyroteo M, Ferreira IA, Elvas
Losonczy H, Siess W. Multiple elec- Djuranovic V, Zrinski Topic R, LB, Ferreira JC, Lapão LV.
trode aggregometry: a new device et al. Association of polymorphisms Remote monitoring systems for
to measure platelet aggregation in coagulation factor genes and patients with chronic diseases in
in whole blood. Thromb Haemost enzymes of homocysteine metabo- primary health care: systematic
2006;96(6):781–8. lism with arterial ischemic stroke in review. JMIR Mhealth Uhealth
72. Nwankwo JO, Gremmel T, Gerrits children. Clin Appl Thromb Hemost 2021;9(12):e28285.
AJ, Mithila FJ, Warburton RR, Hill 2017;23(8):1042–51. 90. Pellman J, Sheikh F. Atrial fibrilla-
NS, et al. Calpain-1 regulates plate- 81. Musso G, Gambino R, Tabibian tion: mechanisms, therapeutics, and
let function in a humanized mouse JH, Ekstedt M, Kechagias S, future directions. Compr Physiol
model of sickle cell disease. Thromb Hamaguchi M, et al. Association 2015;5(2):649–65.
Res 2017;160:58–65. of non-alcoholic fatty liver disease 91. Huang YC, Huang YC, Cheng YC,
73. Subramanian M, Kaplan LJ, Cannon with chronic kidney disease: a sys- Chen M. Choice of antithrombotic
JW. Thromboelastography-guided tematic review and meta-analysis. therapy for patients with atrial fibril-
resuscitation of the Trauma patient. PLoS Med 2014;11(7):e1001680. lation undergoing carotid angio-
JAMA Surg 2019;154(12):1152–3. 82. Guo J, Huang X, Dou L, Yan M, plasty and stenting: a nationwide
74. Da Luz LT, Nascimento B, Shen T, Tang W, et al. Aging and population-based study. Sci Rep
Shankarakutty AK, Rizoli S, aging-related diseases: from molec- 2022;12(1):1417.
Adhikari NK. Effect of thromboe- ular mechanisms to interventions 92. Komen JJ, Pottegård A, Mantel-
lastography (TEG®) and rotational and treatments. Signal Transduct Teeuwisse AK, Forslund T,
thromboelastometry (ROTEM®) Target Ther 2022;7(1):391. Hjemdahl P, Wettermark B, et al.
on diagnosis of coagulopathy, 83. Capranzano P, Angiolillo DJ. Oral anticoagulants in patients with
transfusion guidance and mortality Antithrombotic management of atrial fibrillation at low stroke risk: a
in trauma: descriptive systematic elderly patients with coronary artery multicentre observational study. Eur
review. Crit Care 2014;18(5):518. disease. JACC Cardiovasc Interv Heart J 2022;43(37):3528–38.
75. Tang YD, Wang W, Yang M, 2021;14(7):723–38. 93. Chao TF, Liu CJ, Wang KL, Lin
Zhang K, Chen J, Qiao S, et al. 84. Yang H, Guo R, Li S, Liang F, Tian YJ, Chang SL, Lo LW, et al. Using
Randomized comparisons of dou- C, Zhao X, et al. Systematic analysis the CHA2DS2-VASc score for
ble-dose clopidogrel or adjunctive of gut microbiota in pregnant women refining stroke risk stratification
cilostazol versus standard dual anti- and its correlations with individ- in ‘low-risk’ Asian patients with
platelet in patients with high post- ual heterogeneity. NPJ Biofilms atrial fibrillation. J Am Coll Cardiol
treatment platelet reactivity: results Microbiomes 2020;6(1):32. 2014;64(16):1658–65.
of the CREATIVE trial. Circulation 85. Wu J, Zhang Y, Liao X, Lei Y. 94. Chen JY, Zhang AD, Lu HY, Guo J,
2018;137(21):2231–45. Anticoagulation therapy for non- Wang FF, Li ZC. CHADS2 versus
76. Alseekh S, Aharoni A, Brotman Y, valvular atrial fibrillation: a mini- CHA2DS2-VASc score in assess-
Contrepois K, D’Auria J, Ewald review. Front Med (Lausanne) ing the stroke and thromboembo-
J, et al. Mass spectrometry-based 2020;7:350. lism risk stratification in patients
metabolomics: a guide for anno- 86. Luna DJ, Pandian NKR, Mathur T, with atrial fibrillation: a system-
tation, quantification and best Bui J, Gadangi P, Kostousov VV, atic review and meta-­ analysis.
reporting practices. Nat Methods et al. Tortuosity-powered micro- J Geriatr Cardiol 2013;10(3):
2021;18(7):747–56. fluidic device for assessment of 258–66.
M. Yuan et al., Precision Monitoring of Antithrombotic Therapy in Cardiovascular Disease 19

95. Lane DA, Lip GY. Use of the hemodialysis: a multicenter rand- with aortic valvular stenosis: from
CHA(2)DS(2)-VASc and HAS- omized controlled trial. Circulation new mechanisms to new therapies.
BLED scores to aid decision making 2022;146(23):1735–45. Thromb Res 2016;139:85–9.
for thromboprophylaxis in nonval- 102. Chao TF, Lin YJ, Chang SL, 108. Cheng A, Malkin C, Briffa NP.
vular atrial fibrillation. Circulation Lo LW, Hu YF, Tuan TC, et al. Antithrombotic therapy after heart
2012;126(7):860–5. R2CHADS2 score and thromboem- valve intervention: review of
96. Cameli M, Mandoli GE, Loiacono bolic events after catheter ablation mechanisms, evidence and current
F, Sparla S, Iardino E, Mondillo of atrial fibrillation in comparison guidance. Heart 2023;110:87–93.
S. Left atrial strain: a useful index with the CHA2DS2-VASc score. 109. Lv J, Zhang B, Ye Y, Li Z, Wang
in atrial fibrillation. Int J Cardiol Can J Cardiol 2014;30(4):405–12. W, Zhao Q, et al. Assessment of
2016;220:208–13. 103. Yang Y, Wang Z, Chen Z, Wang X, cardio-renal-hepatic function in
97. Tripathi MM, Egawa S, Wirth Zhang L, Li S, et al. Current status patients with valvular heart dis-
AG, Tshikudi DM, Van Cott EM, and etiology of valvular heart dis- ease: a multi-biomarker approach-
Nadkarni SK. Clinical evalua- ease in China: a population-based the cardio-renal-hepatic score.
tion of whole blood prothrombin survey. BMC Cardiovasc Disord BMC Med 2023;21(1):257.
time (PT) and international nor- 2021;21(1):339. 110. Potter E, Marwick TH. Assessment
malized ratio (INR) using a Laser 104. Harky A, Botezatu B, Kakar S, Ren of left ventricular function by echo-
Speckle Rheology sensor. Sci Rep M, Shirke MM, Pullan M. Mitral cardiography: the case for rou-
2017;7(1):9169. valve diseases: Pathophysiology tinely adding global longitudinal
98. Wimmer NJ, Scirica BM, Stone and interventions. Prog Cardiovasc strain to ejection fraction. JACC
PH. The clinical significance of Dis 2021;67:98–104. Cardiovasc Imaging 2018;11(2 Pt
continuous ECG (ambulatory 105. Otto CM, Nishimura RA, Bonow 1):260–74.
ECG or Holter) monitoring of the RO, Carabello BA, Erwin JP 3rd, 111. Vinding NE, Butt JH, Lauridsen
ST-segment to evaluate ischemia: Gentile F, et al. 2020 ACC/AHA MD, Kristensen SL, Johnsen SP,
a review. Prog Cardiovasc Dis guideline for the management Krøll J, et al. Long-term incidence
2013;56(2):195–202. of patients with valvular heart of ischemic stroke after transient
99. Lip GYH, Keshishian AV, Zhang disease: executive summary: a ischemic attack: a nationwide study
Y, Kang A, Dhamane AD, Luo report of the American College from 2014 to 2020. Circulation
X, et al. Oral anticoagulants for of Cardiology/American Heart 2023;148(13):1000–10.
nonvalvular atrial fibrillation in Association Joint Committee 112. Shah N, Madhavan MV, Gray WA,
patients with high risk of gastro- on Clinical Practice Guidelines. Brener SJ, Ahmad Y, Lindenfeld
intestinal bleeding. JAMA Netw Circulation 2021;143(5):e35–71. J, et al. Prediction of death or HF
Open 2021;4(8):e2120064. 106. Singer DE, Chang Y, Fang MC, hospitalization in patients with
100. Boriani G, Laroche C, Diemberger Borowsky LH, Pomernacki NK, severe FMR: the COAPT risk
I, Popescu MI, Rasmussen LH, Udaltsova N. Should patient charac- score. JACC Cardiovasc Interv
Petrescu L, et al. Glomerular fil- teristics influence target anticoagu- 2022;15(19):1893–905.
tration rate in patients with atrial lation intensity for stroke prevention 113. Cavalcante JL, Lalude OO,
fibrillation and 1-year outcomes. in nonvalvular atrial fibrillation?: Schoenhagen P, Lerakis S.
Sci Rep 2016;6:30271. the ATRIA study. Circ Cardiovasc Cardiovascular magnetic reso-
101. Pokorney SD, Chertow GM, Qual Outcomes 2009;2(4): nance imaging for structural and
Al-Khalidi HR, Gallup D, Dignacco 297–304. valvular heart disease interven-
P, Mussina K, et al. Apixaban for 107. Natorska J, Mazur P, Undas A. tions. JACC Cardiovasc Interv
patients with atrial fibrillation on Increased bleeding risk in patients 2016;9(5):399–425.

You might also like