SYSTEMATIC REVIEW

Biomarkers in atrial fibrillation: an overview J. A. Vılchez,1,2 V. Roldan,3 D. Hernandez-Romero,1 M. Valdes,1 G. Y. H. Lip,4 F. Marın1 Linked Comment: Lip. Int J Clin Pract 2014; 68: 408–9.

1

Department of Cardiology, Hospital Universitario Virgen de la Arrixaca, University of Murcia, Murcia, Spain 2 Department of Clinical Analysis, Hospital Universitario Virgen de la Arrixaca, University of Murcia, Murcia, Spain 3 Hematology and Medical Oncology Unit, Hospital Universitario Morales Meseguer, University of Murcia, Murcia, Spain 4 University of Birmingham Centre for Cardiovascular Sciences, City Hospital, Birmingham, UK Correspondence to: Francisco Marın, MD, PhD, Department of Cardiology, Hospital Universitario Virgen de la Arrixaca, Universidad de Murcia, Ctra Madrid-Cartagena s/n, Murcia, 30120, Spain Tel.: 0034 968398115 Fax: 0034 968369662 Email: [email protected] Disclosure None declared in relation to this manuscript for all authors. VR has received funding for consultancy and lecturing from Bristol-Myers-Squibb, Bayer and Boehringer Ingelheim. FM has received funding for research, consultancy and lecturing from Abbott, Boston Scientifics, Bayer, Astra Zeneca, DaiichiSankyo, BMS/Pfizer and Boehringer Ingelheim. GYHL has served as a consultant for Bayer, Astellas, Merck, AstraZeneca, Sanofi-Aventis, Aryx, Portola, Biotronic, and Boehringer-Ingelheim and has been on the speaker bureau for Bayer, Boehringer-Ingelheim, and Sanofi-Aventis.

434

SUMMARY

Review criteria

Atrial fibrillation (AF) confers a raised risk of stroke and death, and this risk of adverse events is increased by the coexistence of other cardiovascular risk factors. The pathophysiology of AF is complex, involving the role of inflammation, structural remodelling with apoptosis, inflammation or fibrosis. These changes confer a prothrombotic or hypercoagulable state in this arrhythmia. Despite being easy to use for decision-making concerning oral anticoagulant therapy in AF, clinical risk scores used for stratification have shown modest capability in predicting thromboembolic events, and biomarkers may improve our identification of ‘high risk’ patients. Biomarkers, whether measured in the peripheral blood, urine or imagingbased may improve our knowledge of the pathophysiology of AF. Importantly these biomarkers could help in the assessment of AF prognosis. The aim of this review was to summarise the published data about biomarkers studied in AF, with focus on data from randomised prospective clinical trials and large communitybased cohorts. We will also review the application of these biomarkers to prognosis on the main schemes used to help stratify risk in AF.

Introduction Atrial fibrillation (AF) is the most common cardiac arrhythmia, which is associated with increased morbidity and mortality (1). AF increases the risk of stroke and death, and this risk of adverse events is increased by the coexistence of other cardiovascular risk factors such as heart failure, hypertension, diabetes mellitus or prior thromboembolism (2). The pathophysiology of AF is complex and multifactorial, involving ageing and a structural remodelling whereby apoptosis, inflammation and fibrosis are the hallmarks (3). Indeed, left ventricular dysfunction and elevated ventricular filling pressures contribute to atrial remodelling, and the role of the pulmonary veins as one of the key trigger sites for the onset of AF has also been described (4–6). The pathways underlying thrombogenesis in AF are complex. Abnormal changes are consistent with a prothrombotic or hypercoagulable state in AF (7). More specifically, inflammation seems to play an important role in the prothrombotic state associated with AF (8). Furthermore, in recent years there are different data that highlight the association between inflammation to AF itself and AF-related complications (8). Indeed, multiple reports suggest the role of inflammation is not only as a marker of incident AF,

We have reviewed in Pubmed using the following key words: Atrial fibrillation, biomarkers, NTproBNP, Troponin, C-reactive protein, von Willebrand factor, D-dimer, adipokines, renal biomarkers. We have also reviewed out-standing revision manuscript in atrial fibrillation and biomarkers.

Message for the clinic Biomarkers could give us important information about underlying mechanisms and more importantly prognostic information.

but also as an underlying mechanism involved in the induction of AF (9,10).

Risk stratification of AF: the use of clinical risk scores Oral anticoagulation (OAC) is highly effective in reducing stroke risk and mortality rates in patients with AF, but also increases the risk of bleeding (11). To aid decision-making for thromboprophylaxis with OAC, several risk stratification schemes have been developed using clinical characteristics, the most popular being the CHADS2 (congestive heart failure, hypertension, age ≥ 75 years, diabetes mellitus, and prior stroke or transient ischaemic attack) (12). Because of several limitations (13), the CHA2DS2VASc [Cardiac failure or dysfunction, Hypertension, Age ≥ 75 years (Doubled), Diabetes, Stroke (Doubled)–Vascular disease, Age 65–74 years and Sex category (Women)] has been proposed to complement the CHADS2 score (14), reflecting a risk factor-based approach to thromboprophylaxis (15,16). The CHA2DS2-VASc score seems better at identifying the truly low risk group of patients who have an annual stroke risk < 1% (12,14,17). In the view of some authors, CHA2DS2-VASc seems to be overinclusive, categorising a very high percentage of ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443. doi: 10.1111/ijcp.12304

Biomarkers in atrial fibrillation

subjects to be OAC eligible, being exposed to an increased risk of major bleeding (18). The alternative would be to leave some patients untreated (or only given aspirin, which is ineffective and not any safer in terms of bleeding risk) and exposed to the risk of fatal and devastating strokes. Indeed, we recently demonstrated how CHA2DS2-VASc score predicted adverse events beyond thromboembolic risk in AF patients taking OAC (19). Stroke risk is also closely related to bleeding risk, and OAC therapy needs to weigh the benefit from stroke prevention against the bleeding risk. Many thromboembolic risk factors have also been identified as bleeding risk factors (e.g., advanced age or uncontrolled hypertension) (20). The HAS-BLED [(Hypertension, Abnormal renal/liver function, Stroke, Bleeding history or predisposition, Labile International Normalised Ratio, Elderly, Drugs/alcohol concomitantly)] has recently been proposed as a practical tool to assess the individual bleeding risk of real-world AF patients (21,22). The HAS-BLED score may also give important prognostic information regarding death and cardiovascular events, well as bleeding risk although HAS-BLED is a much better predictor of bleeding (23). Clinical risk scores have shown modest capability in predicting thromboembolic events, with low values for area under the curve (14,24,25), with C-statistics between 0.549 and 0.638 (14). Various studies have highlighted the incorporation of biomarkers to improve the prediction power of these scores, enhancing the identification of ‘high risk’ patients who sustain thromboembolism (Table 1) (26–29). Blood-based biomarkers, could improve our knowledge of the pathophysiology of AF (Figure 1). The study of biomarkers related to the different pathways involved in AF (myocardial injury, wall stress or inflammatory markers) could be related to clinical and echocardiographic risk factors of thromboembolism. The aim of this review was to summarise the published data about biomarkers studied in AF, with focus on data from randomised prospective clinical trials and large community-based cohorts. We will also review the application of these biomarkers in prognosis on the main schemes used to stratify the AF risk.

Cardiac biomarkers in AF Myocardial injury Cardiac troponins are contractile proteins of cardiomyocytes and are released during myocardial necrosis, and thus they are known as sensitive and specific biomarkers of myocardial injury (30). Slight elevaª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

tions in troponin levels (troponin T or I) are observed in patients with stable coronary artery disease, heart failure, and also in elderly apparently healthy individuals and have been associated with worse outcomes and increased mortality independent of conventional major coronary risk factors (30–32). The first study that reported circulating troponin I (hsTnI) levels were associated with mortality and major adverse cardiac events in AF was in a cohort of hospitalised patients (33). In a substudy from the RE-LY trial, raised levels of TnI could frequently be detected in patients with AF and risk factors for stroke, and TnI elevation was linked to an increased risk of stroke and mortality (26). Moreover, risk assessment for cardiovascular death, was independently improved when TnI was added to CHADS2 and CHA2DS2-VASc risk scores. These results were confirmed in the study by Roldan et al. (29), in a stable and chronic anticoagulated AF cohort, whereby increased plasma hsTnT levels were associated with an adverse prognosis in AF patients, with regard to cardiovascular events and mortality. Recently, these data were also confirmed in a substudy of the ARISTOTLE trial (27,34). Perhaps troponin increase (hsTnI or hsTnT) is because of AF per se, or caused by coexistent cardiovascular risk factors, or troponin may simply reflect a ‘sick heart’. Thus, there is no established explanation for the association between high troponin and stroke.

Myocyte wall stress B-type natriuretic peptide (BNP) and the stable Nterminal portion of the prohormone, pro-BNP (NTproBNP) are peptides synthesised by myocytes, predominantly in the left ventricle, in response to elevated wall stress. High natriuretic peptide levels correlate positively with cardiac filling pressures, making them excellent markers for abnormal LV wall stress and thus they have been proposed as a marker of LV dysfunction (35). BNP was an independent predictor of new-onset AF in ST segment elevation myocardial infarction patients (36) elevated NTproBNP levels independently predict an increased risk of development of AF (37). A substudy of the RE-LY trial (26), found that NTproBNP was predictive of thromboembolic events and cardiovascular mortality, and even after adjustment by potential confounding factors, the risk of stroke or systemic embolism was doubled to fivefold higher for cardiovascular mortality, in patients with the highest quartiles of NT-proBNP. Similar results have been shown in the ARISTOTLE trial (28), which showed improved risk stratification with NT-proBNP, doubling the risk of stroke and cardiac death.

435

2011

2013 2009 2012

2013 2013

Roldan et al. (56) (composite of cardiovascular events)

Hernandez-Romero et al. (75) Go et al. (82)

Hohnloser et al. (stroke/systemic embolism) (84)

Roldan et al. (83) Hijazi et al. (28)

Hijazi et al. (26) (Results for Vascular death)

2012

2012

Roldan et al. (29)

End-point: mortality

2012

Hijazi et al. (26)

End-point: composite of thromboembolic outcomes

5.07 (2.95–8.71), p < 0.0001

0.705 0.406 0.098 0.006 0.0001

NT-proBNP (> 1402 ng/l)

= = = =
1250 ng/l)

(1.13–1.71) (1.29–1.85) (0.55–1.14), (0.39–0.94), (0.39–1.05), (1.11–1.83), (1.73–2.82),

– NS NS 1.39 1.54 0.79 0.61 0.64 1.42 2.21

D-dimer

vWF (≥ 221 IU/dl)

hsIL6 (≥ 3.35 pg/ml)

1.67 (1.09–2.56), p = 0.019 1.68 (1.10–2.57), p = 0.017 1.97 (1.29–3.02), p = 0.002 1.89 (1.23–2.89), p = 0.004 2.71(1.78–4.13), p < 0.001

hsTnT (≥ 8.04 pg/ml)

2.79 (1.96–3.98), p < 0.0001

0.032 0.023 0.276 0.295 0.001

NT-proBNP (> 1402 ng/l)

= = = =
1250 ng/l)

2013

2.37 2.44 1.46 1.45 2.96 2.35 (1.62–3.40), p < 0.0001

Adiponectin (< 4444 ng/ml) (female patients)

2013

Hernandez-Romero et al. (cardiovascular events) (75) Hijazi et al. (28)

hsIL6 (≥ 3.35 pg/ml)

hsTnT (≥ 8.04 pg/ml)

2012

2.09 (1.22–3.58), p < 0.0001

NT-proBNP (> 1402 ng/l)

Roldan et al. (29)

CHADS2 CHA2DS2-VASc CHADS2 CHA2DS2-VASc CHADS2 CHA2DS2-VASc CHADS2 CHA2DS2-VASc –

Adjusted score

1.68 (0.97–2.89), p = 0.0232

HR 95% CI (multivariate analysis, p-value)

Troponin I (≥ 0.040 lg/l)

Biomarker evaluated

2012

Year

Hijazi et al. (26)

End-point: stroke/TIA

Study

Table 1 Examples of studies linking biomarkers to AF prognosis in several types of events

End-point included in composite thromboembolic outcome

239, p < 0.0001 152, p < 0.0001 211, p < 0.0001 125, p < 0.0001 4.4, p < 0.001 3.7, p < 0.001 2.2, p = 0.003 2.1, p = 0.001 12, p < 0.001 10.3, p < 0.001 – – – – – – – – – 162, p < 0.0001

47, p < 0.0001

70, p = 0.0398 59, p = 0.0492 76, p = 0.1157 63, p = 0.2393 – – – – 12.8, p < 0.001

Relative IDI (%), p-value

436 Biomarkers in atrial fibrillation

ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

2012

2011

2013

2012

2013 2013

Roldan et al. (29)

Roldan et al. (56)

Hernandez-Romero et al. (75) Hermida et al. (44)

Hohnloser et al. (84)

Roldan et al. (83) Hijazi et al. (28)

2011

2012

2013 2013

Roldan et al. (56)

Hohnloser et al. (84)

Roldan et al. (83) Hijazi et al. (28)

< = = = = =
1250 ng/l)

D-dimer

vWF (≥ 221 IU/dl)

4.47 – – NS 0.50 0.48 0.65 1.44 1.07

(0.38–0.66), (0.37–0.64), (0.47–0.91), (1.08–1.94), (0.82–1.40),

p p p p p

= = = = =

0.030 0.004 0.775 0.015 0.0667

(1.86–10.75), p = 0.001

1.28 (0.87–1.88), p < 0.5272

p p p p p p p

p p p p p

NT-proBNP (> 1402 ng/l)

(1.49–4.25), (1.09–4.57), (0.70–1.05), (0.63–0.96), (0.79–1.26), (1.13–1.91), (1.80–2.81),

(1.13–2.83), (1.25–3.20), (1.60–3.85), (1.36–3.52), (1.24–3.32),

1.89 (1.30–2.75), p < 0.0040

1.79 1.99 2.48 2.19 2.03 – – NS NS 2.52 2.23 0.86 0.78 1.00 1.47 2.25

HR 95% CI (multivariate analysis, p-value)

Troponin I (≥ 0.040 lg/l)

Adiponectin (< 4444 ng/ml) hsCRP (≥ 6 mg/dl) All-cause mortality hsCRP (≥ 6 mg/dl) Cardiovascular mortality Lowest eGFR (≤ 50 ml/min) (Cockcroft–Gault equation) Lowest eGFR (≤ 50 ml/min) (CKD-EPI equation) Lowest Cystatin-eGFR (≤ 50 ml/min) Lowest eGFR (< 30 ml/min/1.73 m²) (MDRD equation) NT-proBNP (> 1250 ng/l)

D-dimer

vWF (≥ 221 IU/dl)

hsIL6 (≥ 3.35 pg/ml)

hsTnT (≥ 8.04 pg/ml)

Biomarker evaluated

NS, No significance of the mulltivariate analysis.

2012

Hijazi et al. (26)

End-point: bleeding

Year

Study

Table 1 Continued

CHADS2 CHA2DS2-VASc CHADS2 CHA2DS2-VASc CHADS2 CHA2DS2-VASc HAS-BLED – – – – – CHA2DS2-VASc

CHADS2 CHA2DS2-VASc CHADS2 CHA2DS2-VASc CHADS2 CHA2DS2-VASc HAS-BLED – – CHADS2 CHADS2 – – – – CHA2DS2-VASc

Adjusted score

– – – – – – 11, p = 0.005 – – – – – –

7.2, p < 0.001 3.1, p < 0.001 1.1, p = 0.025 2.7, p = 0.002 8.5, p < 0.001 7.1, p < 0.001 – – – 0.147, p = 0.002 0.034, p = 0.40 – – – – 270, p < 0.0001

Relative IDI (%), p-value

Biomarkers in atrial fibrillation 437

438

Biomarkers in atrial fibrillation

Figure 1 Different pathways involved in the AF pathophysiology related to various biomarkers

Inflammation biomarkers in AF Abnormal changes in systemic inflammation have been related to prothrombotic indices in AF (38). Whether inflammation is an initiator, a consequence, or merely an association of AF is debatable and the results trying to associate the inflammatory biomarkers with AF, are inconsistent (8). C-reactive protein (CRP), is the usual biomarker linked to inflammation and is predominantly synthesised in hepatocytes as an acute-phase reactant (39). For example, Aviles et al. (40) demonstrated that high CRP levels predicted increased risk of developing AF and this was confirmed in other studies (41,42). In a small study, Conway et al. (43) reported the association between CRP and a composite outcome of stroke and death in AF. The prognostic value of CRP, to all-cause mortality and a composite of ischaemic stroke, myocardial infarction or vascular death, was displayed in a larger cohort based on the Stroke Prevention in Atrial Fibrillation III trial (41). Another recent study (42) showed how high sensitive CRP (hsCRP), a marker of low-grade inflammation, was independently associated with AF in men, but apparently not in women, reflecting an elevate and coexisting state of coronary heart disease in their cohort of men. In a substudy of ARIC cohort, Hermida et al. (44) confirmed the results on hsCRP as a predictor of mortality with significant improvement on the CHADS2 score by addition of this biomarker. Other inflammation markers such as, tumour necrosis factor-a (TNF-a), interleukins, monocyte chemoattractan protein-1 (MCP-1) have also been related to AF.

Tumour necrosis factor-a, a pleiotropic proinflammatory molecule, is synthesised mainly by monocytes and macrophages, and is elevated in valvular AF (45) and persistent AF (10). Interleukin-2 (IL2), produced by activated T lymphocytes, is associated with reduced incidence of post-operative AF (46). Another interleukin, IL8 has also shown to be elevated in permanent AF (47). In contrast, MMCP-1, which is a human CC chemokine, has been not associated independently with AF (48,49). A novel biomarker related to inflammation, osteoprotegerin has been related to cardiovascular diseases or atherosclerosis, and Nyrnes et al. (42) found a significant association with AF only on the univariate analysis, and white blood cells in the upper quartile had increased risk of AF (42). The inflammation marker best related to AF, has been interleukin-6 (IL6), a circulating cytokine produced by monocytes, macrophages, T lymphocytes and endothelial cells that can induce a prothrombotic state (50). We have demonstrated raised levels of IL6 in AF, which suggest the presence of an inflammatory state, although this fact appears to be related to clinical variables of the patients, rather than to the presence of AF per se (51). Importantly, IL6 concentrations had implications for prognosis (43). Our recent prognostic study by Roldan et al. (29) was the first to show how hsIL6 levels provided prognostic information that was complementary to clinical risk scores for prediction of long-term cardiovascular events and death, suggesting that hsIL6 and hsTnT may potentially be used to refine clinical risk stratification in AF. ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

Biomarkers in atrial fibrillation

Biomarkers related to prothrombotic state in AF Atrial fibrillation provides abnormal changes in flow, evidenced by stasis in the left atrium. Moreover, abnormal changes in vessel walls include progressive atrial dilatation, endocardial denudation, and oedematous or fibroelastic infiltration of the extracellular matrix. Additionally, abnormal changes in blood constituents are well described in AF, and include haemostatic and platelet activation, as well as inflammation and growth factor changes. These changes fulfil Virchow’s triad for thrombogenesis, and are consistent with a prothrombotic or hypercoagulable state in AF (7). Of note, abnormal concentrations of prothrombotic indices (e.g., prothrombin fragments 1 and 2 and thrombin-antithrombin complexes) are more prominent in patients with stroke who have AF (52). Patients with atrial flutter and impaired left atrial appendage function (as shown by pulsed-wave Doppler) have increased amounts of D-dimer (DD) and b-thromboglobulin (53). Indeed, DD (a fibrin degradation product) has also been shown to predict subsequent thromboembolic events in patients with non-valvular AF, even in those already receiving treatment with warfarin (54,55). However, Roldan et al. (56) did not find that DD levels in an anticoagulated AF cohort were related to prognosis, in contrast to other studies with prognostic value of DD for stroke (57,58). D-dimer as a marker of fibrin turnover, is essentially an index of thrombogenesis, which is raised along with clinical risk factors for thromboembolism (59). More recent trial substudies (RE-LY or ARISTOTLE) described an association between DD levels and the risk of stroke, cardiovascular death and major bleeding outcomes independent of established risk factors including the CHADS2 variables (27). The risk increased with higher DD levels as evidenced by a threefold increase of stroke or systemic embolism and 3.5-fold increase for cardiovascular mortality. These results were confirmed in the ARISTOTLE substudy, which showed how DD levels at baseline, regardless OAC, were related to stroke, mortality and major bleeding (27).

Biomarkers of endothelial damage/ dysfunction in AF Abnormal haemostasis and coagulation are well described in AF and further insights into the hypercoagulable state in AF are evident (7). Plasma levels of soluble E-selectin (sE-sel), von Willebrand factor (vWF) and soluble thrombomodulin (sTM) have been used as indexes of endothelial activation, damª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

age/dysfunction and endothelial damage, respectively. A soluble form of thrombomodulin (sTM) is a recognised marker of endothelial dysfunction and may contribute to the hypercoagulable state in AF. Plasma sTM levels are lower in patients with persistent AF (60), but the biomarker best linked to prognosis in AF has been vWF, which is an established biomarker of endothelial damage/dysfunction with increased plasma levels observed in inflammatory and atherosclerotic vascular diseases, perhaps reflecting a damaged endothelium (61). Furthermore, plasma vWF levels have been associated with independent risk factors for stroke (heart failure, previous stroke, age and diabetes) and stroke risk stratification schemes (43,62). Plasma vWf levels also refined clinical CHADS2 risk stratification scheme for stroke and vascular events among AF patients (63). In addition, inflammatory (58,64) and prothrombotic (57) markers have been related to prognosis in AF patients, even in anticoagulated patients. These data were confirmed by Roldan et al. (56) whereby an increased plasma vWF levels were associated with adverse prognosis in ‘real life’ AF patients, particularly cardiovascular (mainly thrombotic) events, mortality and major bleeding. The addition of vWF as a biomarker risk factor helped to refine these clinical risk stratification schemes for stroke and bleeding.

Platelets in AF Many abnormal changes in platelets seen in AF, could simply indicate underlying vascular comorbidities (7). For example, Choudhury et al. (65) showed higher levels of platelet microparticles and soluble Pselectin in AF patients compared with healthy controls in sinus rhythm, but no difference was seen between patients with AF and disease-matched controls in sinus rhythm, implying the effect was more related to the underlying comorbidities. Increased amounts of b-thromboglobulin, a platelet-specific protein that indicates platelet activation and is released from a-granules during platelet aggregation and subsequent thrombus formation, have been shown in patients with both valvular and non-valvular AF compared with controls in sinus rhythm (7,66). Increased levels of CD62P (P-selectin) expression on platelets and platelet–leucocyte conjugates, could predispose to thrombosis and vascular events (67). Indeed, the Rotterdam study (68) showed how plasma concentrations of soluble P-selectin were predictive of adverse clinical outcomes in elderly patients with AF. Furthermore, Hayashi et al. (69) demonstrated how acute induction of AF significantly increased the expression of P-selectin on platelets and microparticles. Other recent works illustrates

439

440

Biomarkers in atrial fibrillation

the possible prothrombotic behaviour of microparticles (65,70) in AF.

Adipokines in AF Adipokines may be related to incident AF through several pathways, involving inflammation or through AF risk factors such as obesity and heart failure (71). Resistin has been associated with increased insulin resistance and has proinflammatory, prohypertrophic effects (72). Adiponectin presents anti-inflammatory, atherogenic and antihypertrophic functions (73) and both of them has been associated with multiple known risk factors for AF, including inflammation, diabetes, obesity, myocardial infarction and incident heart failure (71). Several recent studies have linked adipokines with prognosis in AF, and one reported how high concentrations of adiponectin were related to persistent AF (74). Another study by Rienstra et al., found higher concentrations of resistin were associated with incident AF, thus supporting the role of inflammation in AF initiation, but the relation was attenuated by adjustment for CRP. Of note, they did not detect a statistically significant association between adiponectin and incident AF (71). In contrast, we recently reported data on adiponectin as a prognostic biomarker in AF (75). Adiponectin levels have been related to different atherosclerotic risk factors (76,77), and AF may be indicative of advanced atherosclerosis (78). Our previous study found how low levels of adiponectin were independently associated with adverse cardiovascular events but only in female AF patients (75) and the lack of association in men could be because of testosterone decreasing adiponectin production (79). Our data confirmed the importance of AF as a risk marker of atherosclerotic vascular damage and adiponectin could exert a protective role against cardiovascular diseases.

Renal function biomarkers in AF Atrial fibrillation is usual in patients with chronic kidney disease (CKD) at different stages of severity, commonly in end renal stage, and this prevalence is increased in elderly populations (80). The prevalence of AF increases with a decrease in the estimated glomerular filtration rate (eGFR) (81), and conversely, CKD increases the risk of thromboembolism in AF independently of other risk factors (82). Relating renal function with final outcomes, Go et al. reported an independent stroke risk increase with reduced eGFR or if proteinuria was present (82). Roldan et al. (83) who showed a decreased

eGFR > 10 ml/min/1.73 m2 in 21% of patients, with a 1/5 of followed-up patients developing severe CKD (≤ 30 ml/min/1.73 m2). This study also showed that the presence of impaired renal function was also associated consistently with the development of adverse cardiovascular events, mortality and bleeding, even after adjusting for the CHADS2 score. In the ARISTOTLE trial cohort, there were increased rates of stroke and bleedings using warfarin regardless the eGFR calculate by the Cockcroft–Gault and Chronic Kidney Disease Epidemiology Collaboration equations (CKD-EPI) (84). Renal impairment constitutes a major risk factor for thromboembolic and cardiovascular events in AF (85). Severe renal dysfunction is not included in neither of the two stroke risk stratification scores, but it has been proposed that CKD or proteinuria could be included to CHA2DS2-VASc, being the little ‘c’ letter indicating ‘chronic severe renal impairment’ (85). Unfortunately, to properly test this hypothesis, stroke risk should be validated in non-anticoagulated populations. The recent study published by Roldan et al., based on c-statistics and the integrated discrimination improvement, shows that adding CKD to the CHADS2 and CHA2DS2-VASc stroke risk scores did not independently improve the predictive value of current clinical risk scores (86). A new reliable considered markers of renal function, Cystatin C and b-Trace protein, have also been considered biomarkers who reflect microvascular renal dysfunction (87,88). Cystatin C has also been studied related to AF in the study by Hohnloser et al. (84). They showed how high levels of Cystatin C, when added to the eGFR equation, were associated with increased HR rates of stroke or systemic embolism, mortality and major bleeding in patients taking warfarin (84).

Genetic polymorphisms in AF Polymorphisms associated with AF have shown relatively large risk estimates. The robustness of such estimates across populations and study designs have been recently showed in an interesting meta-analysis by Smith et al. (89). However, limited data support the use of SNPs in risk stratification of stroke. Several polymorphisms have been studied and highlight those related to fibrinogen and platelets (90). The FGA T331A polymorphism (rs6050), results in a change in the a-fibrinogen gene by a threonine to alanine amino acid substitution at position 331 (T331A). This polymorphism was studied by Carter et al. (91) and showed an association with an ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

Biomarkers in atrial fibrillation

increased mortality in patients with stroke and AF compared with those in sinus rhythm, suggesting that possession of A331 might lead to an increased susceptibility for embolisation of thrombus, possibly because of defective FXIII-dependent cross-linking. The FGB g.4577G >A polymorphism (rs1800790), is located in the promoter region of the b-fibrinogen gene. Hyperfibrinogenemia is one important risk factor for cardiovascular disease and stroke (92) and this polymorphism has been related to coronary atherosclerotic disease (93). Bozdemir et al. (94), found that the b-fibrinogen polymorphism was associated with the development of left atrial thrombus or spontaneous echo contrast, suggesting that b-fibrinogen g.4577G> A polymorphism could be a marker for the prediction of thromboembolism risk in patients with AF. Marın et al. (95) reported that F13A1V34L polymorphism was independently associated with the prothrombotic and inflammatory state in AF patients, as evidenced by its association with raised tissue factor and IL6 levels, but not with platelet activation. Roldan et al. (96) also tested the possible role of Factor 7 polymorphism on AF thromboembolic risk, showing a lower prevalence of the F7 g.4727_4728ins10 (rs5742910) polymorphism amongst patients without stroke compared with patients with stroke. Of the polymorphisms associated with platelet functionality, the integrin a2 gene (ITGA2) has had interesting results. This gene is located on chromosome 5q11.2, and the silent change in the coding region at position g.67214C>T (rs1126643) has a correlation with platelet GPIa/IIa density (97). Thus, subjects with the T allele have an increased potential

References 1 Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent risk factor for stroke: the Framingham Study. Stroke 1991; 22(8): 983–8. 2 Stroke Risk in Atrial Fibrilation Working Group. Independent predictors of stroke in patients with atrial fibrillation: a systematic review. Neurology 2007; 69(6): 546–54. 3 Daoud EG, Bogun F, Goyal R, Harvey M, Man KC, Strickberger SA, et al. Effect of atrial fibrillation on atrial refractoriness in humans. Circulation 1996; 94(7): 1600–6. 4 Haissaguerre M, Jais P, Shah DC, Takahashi A, Hocini M, Quiniou G, et al. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Med 1998; 339(10): 659–66. 5 Savelieva I, John CA. Atrial fibrillation and heart failure: natural history and pharmacological treatment. Europace 2004; 5(Suppl. 1): S5–19. 6 Tsang TS, Gersh BJ, Appleton CP, Tajik AJ, Barnes ME, Bailey KR, et al. Left ventricular diastolic dysfunction as a predictor of the first diagnosed non-

ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

7

8

9

10

11

12

441

of platelet adhesion and a tendency to arterial thrombosis.

Conclusions Biomarkers could be useful in refine clinical risk stratification scores, to identify patients at high risk for stroke and thromboembolism. Biomarkers may also increase our knowledge on AF pathogenesis. Several markers reflect the pathophysiologic process for development of AF, while others may simply be suited as markers of risk for future cardiovascular events. Another point of view, is the possibility for a multimarker strategy that will improve better overall risk stratification, as with coronary artery disease (98) or acute coronary syndrome (99). Also, these biomarkers could serve as indices of ongoing thrombogenesis, to test antithrombotic regimens and help decision-making on dose selection.

Funding This work was partially supported by Sociedad Espa~ nola de Cardiologıa, RD06/0014/039, (RECAVA) from ISCIII, Beca Cajamurcia-FFIS 2010; and PI11/ 00566-FEDER from ISCIII.

Acknowledgements JA Vı´lchez holds a research grant ‘Rıo Hortega’ by the Instituto de Salud Carlos III, Madrid, Spain. D Hernandez-Romero holds a postdoctoral position, ‘Sara Borrel’ grant by the Instituto de Salud Carlos III, Madrid, Spain.

valvular atrial fibrillation in 840 elderly men and women. J Am Coll Cardiol 2002; 40(9): 1636–44. Watson T, Shantsila E, Lip GY. Mechanisms of thrombogenesis in atrial fibrillation: Virchow’s triad revisited. Lancet 2009; 373(9658): 155–66. Guo Y, Lip GY, Apostolakis S. Inflammation in atrial fibrillation. J Am Coll Cardiol 2012; 60(22): 2263–70. Rudolph V, Andrie RP, Rudolph TK, Friedrichs K, Klinke A, Hirsch-Hoffmann B, et al. Myeloperoxidase acts as a profibrotic mediator of atrial fibrillation. Nat Med 2010; 16(4): 470–4. Li J, Solus J, Chen Q, Rho YH, Milne G, Stein CM, et al. Role of inflammation and oxidative stress in atrial fibrillation. Heart Rhythm 2010; 7(4): 438–44. Singer DE, Chang Y, Fang MC, Borowsky LH, Pomernacki NK, Udaltsova N, et al. Should patient characteristics influence target anticoagulation intensity for stroke prevention in nonvalvular atrial fibrillation?: the ATRIA study. Circ Cardiovasc Qual Outcomes 2009; 2(4): 297–304. Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from

13

14

15

16

the National Registry of Atrial Fibrillation. JAMA 2001; 285(22): 2864–70. Karthikeyan G, Eikelboom JW. The CHADS2 score for stroke risk stratification in atrial fibrillation– friend or foe? Thromb Haemost 2010; 104(1): 45–8. Lip GY, Nieuwlaat R, Pisters R, Lane DA, Crijns HJ. Refining clinical risk stratification for predicting stroke and thromboembolism in atrial fibrillation using a novel risk factor-based approach: the euro heart survey on atrial fibrillation. Chest 2010; 137(2): 263–72. Camm AJ, Kirchhof P, Lip GY, Schotten U, Savelieva I, Ernst S, et al. Guidelines for the management of atrial fibrillation: the Task Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Eur Heart J 2010; 31(19): 2369–429. Camm AJ, Lip GY, De CR, Savelieva I, Atar D, Hohnloser SH, et al. 2012 focused update of the ESC Guidelines for the management of atrial fibrillation: an update of the 2010 ESC Guidelines for the management of atrial fibrillation – developed with the special contribution of the European Heart Rhythm Association. Europace 2012; 14(10): 1385–413.

442

Biomarkers in atrial fibrillation

17 Olesen JB, Lip GY, Hansen ML, Hansen PR, Tolstrup JS, Lindhardsen J, et al. Validation of risk stratification schemes for predicting stroke and thromboembolism in patients with atrial fibrillation: nationwide cohort study. BMJ 2011; 342: d124. 18 Providencia R, Paiva L, Barra S. Risk stratification of patients with atrial fibrillation: biomarkers and other future perspectives. World J Cardiol 2012; 4 (6): 195–200. 19 Jover E, Roldan V, Gallego P, Hernandez-Romero D, Valdes M, Vicente V, et al. Predictive value of the CHA2DS2-VASc score in atrial fibrillation patients at high risk for stroke despite oral anticoagulation. Rev Esp Cardiol (Engl Ed) 2012; 65(7): 627–33. 20 Lip GY, Andreotti F, Fauchier L, Huber K, Hylek E, Knight E, et al. Bleeding risk assessment and management in atrial fibrillation patients. Executive summary of a position document from the European Heart Rhythm Association [EHRA], endorsed by the European Society of Cardiology [ESC] working group on thrombosis. Thromb Haemost 2011; 106(6): 997–1011. 21 Pisters R, Lane DA, Nieuwlaat R, de Vos CB, Crijns HJ, Lip GY. A novel user-friendly score (HAS-BLED) to assess 1-year risk of major bleeding in patients with atrial fibrillation: the Euro Heart Survey. Chest 2010; 138(5): 1093–100. 22 Lip GY, Frison L, Halperin JL, Lane DA. Comparative validation of a novel risk score for predicting bleeding risk in anticoagulated patients with atrial fibrillation: the HAS-BLED (Hypertension, Abnormal Renal/Liver Function, Stroke, Bleeding History or Predisposition, Labile INR, Elderly, Drugs/Alcohol Concomitantly) score. J Am Coll Cardiol 2011; 57(2): 173–80. 23 Gallego P, Roldan V, Torregrosa JM, Galvez J, Valdes M, Vicente V, et al. Relation of the HAS-BLED bleeding risk score to major bleeding, cardiovascular events, and mortality in anticoagulated patients with atrial fibrillation. Circ Arrhythm Electrophysiol 2012; 5(2): 312–8. 24 van Staa TP, Setakis E, Di Tanna GL, Lane DA, Lip GY. A comparison of risk stratification schemes for stroke in 79,884 atrial fibrillation patients in general practice. J Thromb Haemost 2011; 9(1): 39–48. 25 Fang MC, Go AS, Chang Y, Borowsky L, Pomernacki NK, Singer DE. Comparison of risk stratification schemes to predict thromboembolism in people with nonvalvular atrial fibrillation. J Am Coll Cardiol 2008; 51(8): 810–5. 26 Hijazi Z, Oldgren J, Andersson U, Connolly SJ, Ezekowitz MD, Hohnloser SH, et al. Cardiac biomarkers are associated with an increased risk of stroke and death in patients with atrial fibrillation: a Randomized Evaluation of Long-term Anticoagulation Therapy (RE-LY) substudy. Circulation 2012; 125(13): 1605–16. 27 Hijazi Z, Oldgren J, Siegbahn A, Granger CB, Wallentin L. Biomarkers in atrial fibrillation: a clinical review. Eur Heart J 2013; 34(20): 1475–80. 28 Hijazi Z, Wallentin L, Siegbahn A, Andersson U, Christersson C, Ezekowitz J, et al. N-terminal pro-B-type natriuretic peptide for risk assessment in patients with atrial fibrillation: insights from the ARISTOTLE Trial (Apixaban for the Prevention of Stroke in Subjects with Atrial Fibrillation). J Am Coll Cardiol 2013; 61(22): 2274–84.

29 Roldan V, Marin F, Diaz J, Gallego P, Jover E, Romera M, et al. High sensitivity cardiac troponin T and interleukin-6 predict adverse cardiovascular events and mortality in anticoagulated patients with atrial fibrillation. J Thromb Haemost 2012; 10 (8): 1500–7. 30 Zethelius B, Johnston N, Venge P. Troponin I as a predictor of coronary heart disease and mortality in 70-year-old men: a community-based cohort study. Circulation 2006; 113(8): 1071–8. 31 Omland T, de Lemos JA, Sabatine MS, Christophi CA, Rice MM, Jablonski KA, et al. A sensitive cardiac troponin T assay in stable coronary artery disease. N Engl J Med 2009; 361(26): 2538–47. 32 Horwich TB, Patel J, MacLellan WR, Fonarow GC. Cardiac troponin I is associated with impaired hemodynamics, progressive left ventricular dysfunction, and increased mortality rates in advanced heart failure. Circulation 2003; 108(7): 833–8. 33 van den Bos EJ, Constantinescu AA, van Domburg RT, Akin S, Jordaens LJ, Kofflard MJ. Minor elevations in troponin I are associated with mortality and adverse cardiac events in patients with atrial fibrillation. Eur Heart J 2011; 32(5): 611–7. 34 Wallentin L, Hijazi Z, Siegbahn A, Schollin M, Alexander JH, Atar D, et al. High sensitivity troponin-T for risk stratification in atrial fibrillation during treatment with apixaban or warfarin. Eur Heart J 2012; 33: (Abstract supplement 53). 35 Daniels LB, Maisel AS. Natriuretic peptides. J Am Coll Cardiol 2007; 50(25): 2357–68. 36 Asanin M, Stankovic S, Mrdovic I, Matic D, Savic L, Majkic-Singh N, et al. B-type natriuretic peptide predicts new-onset atrial fibrillation in patients with ST-segment elevation myocardial infarction treated by primary percutaneous coronary intervention. Peptides 2012; 35(1): 74–7. 37 Patton KK, Ellinor PT, Heckbert SR, Christenson RH, DeFilippi C, Gottdiener JS, et al. N-terminal pro-B-type natriuretic peptide is a major predictor of the development of atrial fibrillation: the Cardiovascular Health Study. Circulation 2009; 120 (18): 1768–74. 38 Boos CJ, Anderson RA, Lip GY. Is atrial fibrillation an inflammatory disorder? Eur Heart J 2006; 27(2): 136–49. 39 Devaraj S, Singh U, Jialal I. The evolving role of C-reactive protein in atherothrombosis. Clin Chem 2009; 55(2): 229–38. 40 Aviles RJ, Martin DO, Apperson-Hansen C, Houghtaling PL, Rautaharju P, Kronmal RA, et al. Inflammation as a risk factor for atrial fibrillation. Circulation 2003; 108(24):3006–10. 41 Lip GY, Patel JV, Hughes E, Hart RG. High-sensitivity C-reactive protein and soluble CD40 ligand as indices of inflammation and platelet activation in 880 patients with nonvalvular atrial fibrillation: relationship to stroke risk factors, stroke risk stratification schema, and prognosis. Stroke 2007; 38(4): 1229–37. 42 Nyrnes A, Njolstad I, Mathiesen EB, Wilsgaard T, Hansen JB, Skjelbakken T, et al. Inflammatory biomarkers as risk factors for future atrial fibrillation. An eleven-year follow-up of 6315 men and women: the Tromso study. Gend Med 2012; 9(6): 536–47. 43 Conway DS, Buggins P, Hughes E, Lip GY. Prognostic significance of raised plasma levels of interleukin-6 and C-reactive protein in atrial fibrillation. Am Heart J 2004; 148(3): 462–6.

44 Hermida J, Lopez FL, Montes R, Matsushita K, Astor BC, Alonso A. Usefulness of high-sensitivity C-reactive protein to predict mortality in patients with atrial fibrillation (from the Atherosclerosis Risk In Communities [ARIC] Study). Am J Cardiol 2012; 109(1): 95–9. 45 Qu YC, Du YM, Wu SL, Chen QX, Wu HL, Zhou SF. Activated nuclear factor-kappaB and increased tumor necrosis factor-alpha in atrial tissue of atrial fibrillation. Scand Cardiovasc J 2009; 43(5): 292–7. 46 Hak L, Mysliwska J, Wieckiewicz J, Szyndler K, Siebert J, Rogowski J. Interleukin-2 as a predictor of early postoperative atrial fibrillation after cardiopulmonary bypass graft (CABG). J Interferon Cytokine Res 2009; 29(6): 327–32. 47 Liuba I, Ahlmroth H, Jonasson L, Englund A, Jonsson A, Safstrom K, et al. Source of inflammatory markers in patients with atrial fibrillation. Europace 2008; 10(7): 848–53. 48 Marcus GM, Whooley MA, Glidden DV, Pawlikowska L, Zaroff JG, Olgin JE. Interleukin-6 and atrial fibrillation in patients with coronary artery disease: data from the Heart and Soul Study. Am Heart J 2008; 155(2): 303–9. 49 Schnabel RB, Larson MG, Yamamoto JF, Kathiresan S, Rong J, Levy D, et al. Relation of multiple inflammatory biomarkers to incident atrial fibrillation. Am J Cardiol 2009; 104(1): 92–6. 50 Kerr R, Stirling D, Ludlam CA. Interleukin 6 and haemostasis. Br J Haematol 2001; 115(1): 3–12. 51 Roldan V, Marin F, Blann AD, Garcia A, Marco P, Sogorb F, et al. Interleukin-6, endothelial activation and thrombogenesis in chronic atrial fibrillation. Eur Heart J 2003; 24(14): 1373–80. 52 Turgut N, Akdemir O, Turgut B, Demir M, Ekuklu G, Vural O, et al. Hypercoagulopathy in stroke patients with nonvalvular atrial fibrillation: hematologic and cardiologic investigations. Clin Appl Thromb Hemost 2006; 12(1): 15–20. 53 Sakurai K, Hirai T, Nakagawa K, Kameyama T, Nozawa T, Asanoi H, et al. Left atrial appendage function and abnormal hypercoagulability in patients with atrial flutter. Chest 2003; 124(5): 1670–4. 54 Vene N, Mavri A, Kosmelj K, Stegnar M. High D-dimer levels predict cardiovascular events in patients with chronic atrial fibrillation during oral anticoagulant therapy. Thromb Haemost 2003; 90 (6): 1163–72. 55 Nozawa T, Inoue H, Hirai T, Iwasa A, Okumura K, Lee JD, et al. D-dimer level influences thromboembolic events in patients with atrial fibrillation. Int J Cardiol 2006; 109(1): 59–65. 56 Roldan V, Marin F, Muina B, Torregrosa JM, Hernandez-Romero D, Valdes M, et al. Plasma von Willebrand factor levels are an independent risk factor for adverse events including mortality and major bleeding in anticoagulated atrial fibrillation patients. J Am Coll Cardiol 2011; 57(25): 2496–504. 57 Sadanaga T, Sadanaga M, Ogawa S. Evidence that D-dimer levels predict subsequent thromboembolic and cardiovascular events in patients with atrial fibrillation during oral anticoagulant therapy. J Am Coll Cardiol 2010; 55(20): 2225–31. 58 Maehama T, Okura H, Imai K, Yamada R, Obase K, Saito K, et al. Usefulness of CHADS2 score to predict C-reactive protein, left atrial blood stasis, and prognosis in patients with nonrheumatic atrial fibrillation. Am J Cardiol 2010; 106(4): 535–8.

ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

Biomarkers in atrial fibrillation

59 Habara S, Dote K, Kato M, Sasaki S, Goto K, Takemoto H, et al. Prediction of left atrial appendage thrombi in non-valvular atrial fibrillation. Eur Heart J 2007; 28(18): 2217–22. 60 Freestone B, Chong AY, Nuttall S, Blann AD, Lip GY. Soluble E-selectin, von Willebrand factor, soluble thrombomodulin, and total body nitrate/nitrite product as indices of endothelial damage/dysfunction in paroxysmal, persistent, and permanent atrial fibrillation. Chest 2007; 132(4): 1253–8. 61 Blann AD, Lip GY. The endothelium in atherothrombotic disease: assessment of function, mechanisms and clinical implications. Blood Coagul Fibrinolysis 1998; 9(4): 297–306. 62 Conway DS, Pearce LA, Chin BS, Hart RG, Lip GY. Plasma von Willebrand factor and soluble p-selectin as indices of endothelial damage and platelet activation in 1321 patients with nonvalvular atrial fibrillation: relationship to stroke risk factors. Circulation 2002; 106(15): 1962–7. 63 Lip GY, Lane D, Van WC, Hart RG. Additive role of plasma von Willebrand factor levels to clinical factors for risk stratification of patients with atrial fibrillation. Stroke 2006; 37(9): 2294–300. 64 Ferro D, Loffredo L, Polimeni L, Fimognari F, Villari P, Pignatelli P, et al. Soluble CD40 ligand predicts ischemic stroke and myocardial infarction in patients with nonvalvular atrial fibrillation. Arterioscler Thromb Vasc Biol 2007; 27(12): 2763–8. 65 Choudhury A, Chung I, Blann AD, Lip GY. Elevated platelet microparticle levels in nonvalvular atrial fibrillation: relationship to P-selectin and antithrombotic therapy. Chest 2007; 131(3): 809–15. 66 Kamath S, Chin BS, Blann AD, Lip GY. A study of platelet activation in paroxysmal, persistent and permanent atrial fibrillation. Blood Coagul Fibrinolysis 2002; 13(7): 627–36. 67 Chirinos JA, Castrellon A, Zambrano JP, Jimenez JJ, Jy W, Horstman LL, et al. Digoxin use is associated with increased platelet and endothelial cell activation in patients with nonvalvular atrial fibrillation. Heart Rhythm 2005; 2(5): 525–9. 68 Heeringa J, Conway DS, van der Kuip DA, Hofman A, Breteler MM, Lip GY, et al. A longitudinal population-based study of prothrombotic factors in elderly subjects with atrial fibrillation: the Rotterdam Study 1990-1999. J Thromb Haemost 2006; 4 (9): 1944–9. 69 Hayashi M, Takeshita K, Inden Y, Ishii H, Cheng XW, Yamamoto K, et al. Platelet activation and induction of tissue factor in acute and chronic atrial fibrillation: involvement of mononuclear cell-platelet interaction. Thromb Res 2011; 128(6): e113–8. 70 Montoro-Garcia S, Shantsila E, Marin F, Blann A, Lip GY. Circulating microparticles: new insights into the biochemical basis of microparticle release and activity. Basic Res Cardiol 2011; 106(6): 911–23. 71 Rienstra M, Sun JX, Lubitz SA, Frankel DS, Vasan RS, Levy D, et al. Plasma resistin, adiponectin, and risk of incident atrial fibrillation: the Framingham Offspring Study. Am Heart J 2012; 163(1): 119–24. 72 Filkova M, Haluzik M, Gay S, Senolt L. The role of resistin as a regulator of inflammation: implications

ª 2013 John Wiley & Sons Ltd Int J Clin Pract, April 2014, 68, 4, 434–443

73

74

75

76

77

78

79

80

81

82

83

84

85

86

for various human pathologies. Clin Immunol 2009; 133(2): 157–70. Karmazyn M, Purdham DM, Rajapurohitam V, Zeidan A. Signalling mechanisms underlying the metabolic and other effects of adipokines on the heart. Cardiovasc Res 2008; 79(2): 279–86. Shimano M, Shibata R, Tsuji Y, Kamiya H, Uchikawa T, Harata S, et al. Circulating adiponectin levels in patients with atrial fibrillation. Circ J 2008; 72(7): 1120–4. Hernandez-Romero D, Jover E, Marin F, Vilchez JA, Manzano-Fernandez S, Romera M, et al. The prognostic role of the adiponectin levels in atrial fibrillation. Eur J Clin Invest 2013; 43(2): 168–73. Empana JP. Adiponectin isoforms and cardiovascular disease: the epidemiological evidence has just begun. Eur Heart J 2008; 29(10): 1221–3. Tilg H, Moschen AR. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol 2006; 6(10): 772–83. Heeringa J, van der Kuip DA, Hofman A, Kors JA, van Rooij FJ, Lip GY, et al. Subclinical atherosclerosis and risk of atrial fibrillation: the Rotterdam study. Arch Intern Med 2007; 167(4): 382–7. Nishizawa H, Shimomura I, Kishida K, Maeda N, Kuriyama H, Nagaretani H, et al. Androgens decrease plasma adiponectin, an insulin-sensitizing adipocyte-derived protein. Diabetes 2002; 51(9): 2734–41. Soliman EZ, Prineas RJ, Go AS, Xie D, Lash JP, Rahman M, et al. Chronic kidney disease and prevalent atrial fibrillation: the chronic renal insufficiency cohort (CRIC). Am Heart J 2010; 159(6): 1102–7. Iguchi Y, Kimura K, Kobayashi K, Aoki J, Terasawa Y, Sakai K, et al. Relation of atrial fibrillation to glomerular filtration rate. Am J Cardiol 2008; 102 (8): 1056–9. Go AS, Fang MC, Udaltsova N, Chang Y, Pomernacki NK, Borowsky L, et al. Impact of proteinuria and glomerular filtration rate on risk of thromboembolism in atrial fibrillation: the anticoagulation and risk factors in atrial fibrillation (ATRIA) study. Circulation 2009; 119(10): 1363–9. Roldan V, Marin F, Fernandez H, Manzano-Fernandez S, Gallego P, Valdes M, et al. Renal impairment in a “real-life” cohort of anticoagulated patients with atrial fibrillation (implications for thromboembolism and bleeding). Am J Cardiol 2013; 111(8): 1159–64. Hohnloser SH, Hijazi Z, Thomas L, Alexander JH, Amerena J, Hanna M, et al. Efficacy of apixaban when compared with warfarin in relation to renal function in patients with atrial fibrillation: insights from the ARISTOTLE trial. Eur Heart J 2012; 33 (22): 2821–30. Marinigh R, Lane DA, Lip GY. Severe renal impairment and stroke prevention in atrial fibrillation: implications for thromboprophylaxis and bleeding risk. J Am Coll Cardiol 2011; 57(12): 1339–48. Roldan V, Marin F, Manzano-Fernandez S, Fernandez H, Gallego P, Valdes M, et al. Does chronic kidney disease improve the predictive value of the CHADS2 and CHA2DS2-VASc stroke stratification risk scores for atrial fibrillation? Thromb Haemost 2013; 109(5): 956–60.

443

87 Orenes-Pinero E, Manzano-Fernandez S, LopezCuenca A, Marin F, Valdes M, Januzzi JL. Beta-trace protein: from GFR marker to cardiovascular risk predictor. Clin J Am Soc Nephrol 2013; 8(5): 873–81. 88 Laterza OF, Price CP, Scott MG. Cystatin C: an improved estimator of glomerular filtration rate? Clin Chem 2002; 48(5): 699–707. 89 Smith JG, Almgren P, Engstrom G, Hedblad B, Platonov PG, Newton-Cheh C, et al. Genetic polymorphisms for estimating risk of atrial fibrillation: a literature-based meta-analysis. J Intern Med 2012; 272(6): 573–82. 90 Lopez-Cuenca A, Marin F, Roldan V, Gonzalez-Conejero R, Hernandez-Romero D, Valdes M, et al. Genetic polymorphisms and atrial fibrillation: insights into the prothrombotic state and thromboembolic risk. Ann Med 2010; 42(8): 562–75. 91 Carter AM, Catto AJ, Grant PJ. Association of the alpha-fibrinogen Thr312Ala polymorphism with poststroke mortality in subjects with atrial fibrillation. Circulation 1999; 99(18): 2423–6. 92 Lip GY. Fibrinogen and cardiovascular disorders. QJM 1995; 88(3): 155–65. 93 Yu Q, Safavi F, Roberts R, Marian AJ. A variant of beta fibrinogen is a genetic risk factor for coronary artery disease and myocardial infarction. J Investig Med 1996; 44(4): 154–9. 94 Bozdemir V, Kirimli O, Akdeniz B, Ulgenalp A, Aslan A, Kala V, et al. The association of beta-fibrinogen 455 G/A gene polymorphism with left atrial thrombus and severe spontaneous echo contrast in atrial fibrillation. Anadolu Kardiyol Derg 2010; 10(3): 209–15. 95 Marin F, Corral J, Roldan V, Gonzalez-Conejero R, del Rey ML, Sogorb F, et al. Factor XIII Val34Leu polymorphism modulates the prothrombotic and inflammatory state associated with atrial fibrillation. J Mol Cell Cardiol 2004; 37(3): 699–704. 96 Roldan V, Marin F, Gonzalez-Conejero R, Garcia-Honrubia A, Marti S, Alfaro A, et al. Factor VII -323 decanucleotide D/I polymorphism in atrial fibrillation: implications for the prothrombotic state and stroke risk. Ann Med 2008; 40(7): 553–9. 97 Corral J, Gonzalez-Conejero R, Rivera J, Ortuno F, Aparicio P, Vicente V. Role of the 807 C/T polymorphism of the alpha2 gene in platelet GP Ia collagen receptor expression and function – effect in thromboembolic diseases. Thromb Haemost 1999; 81(6): 951–6. 98 Lindahl B, Toss H, Siegbahn A, Venge P, Wallentin L. Markers of myocardial damage and inflammation in relation to long-term mortality in unstable coronary artery disease. FRISC Study Group. Fragmin during Instability in Coronary Artery Disease. N Engl J Med 2000; 343(16): 1139–47. 99 Tello-Montoliu A, Marin F, Roldan V, Mainar L, Lopez MT, Sogorb F, et al. A multimarker risk stratification approach to non-ST elevation acute coronary syndrome: implications of troponin T, CRP, NT pro-BNP and fibrin D-dimer levels. J Intern Med 2007; 262(6): 651–8.

Paper received July 2013, accepted August 2013

Biomarkers in atrial fibrillation: an overview.

Atrial fibrillation (AF) confers a raised risk of stroke and death, and this risk of adverse events is increased by the coexistence of other cardiovas...
212KB Sizes 0 Downloads 0 Views