You are on page 1of 5

International Journal of Cardiology 240 (2017) 25–29

Contents lists available at ScienceDirect

International Journal of Cardiology

journal homepage: www.elsevier.com/locate/ijcard

Serum uric acid on admission predicts in-hospital mortality in patients


with acute coronary syndrome
Marco Magnoni a, Martina Berteotti a, Ferruccio Ceriotti a, Vincenzo Mallia a, Vittoria Vergani a,
Giovanni Peretto a, Giulia Angeloni b, Nicole Cristell a, Attilio Maseri c, Domenico Cianflone a,⁎,1
a
IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
b
Department of Heart and Vessels, Careggi Hospital, University of Florence, Florence, Italy
c
Heart Care Foundation Onlus, Florence, Italy

a r t i c l e i n f o a b s t r a c t

Article history: Background: Despite the association between uric acid and cardiovascular disease has been known for decades,
Received 9 February 2017 the prognostic value of serum uric acid (UA) in all clinical manifestations of acute coronary syndrome (ACS),
Received in revised form 30 March 2017 namely ST-elevation myocardial infarction (STEMI), NSTEMI and unstable angina, has not been definitively
Accepted 10 April 2017 assessed.
Available online 11 April 2017
Methods: This retrospective analysis included patients from previous SPAI and FAMI studies with the aim to in-
vestigate the association between serum uric acid and major adverse cardiovascular events at 180 days from hos-
Keywords:
Acute coronary syndrome
pital admission.
Uric acid Results: 1548 patients were considered and divided in four groups, according UA concentration. Uricemia was
Biomarkers significantly associated with gender, BMI, arterial hypertension, HDL-cholesterol, triglycerides, metabolic
Prognosis syndrome and glomerular filtration rate in univariate analysis. Multivariate logistic regression indicated that
UA N 6.0 mg/dL on admission increased the risk of in-hospital mortality in overall population (OR 2.9, 95%CI
1.4–6.1; p = 0.0057) and in patients with de novo ACS (OR 3.2, 95%CI 1.5–6.8; p = 0.0033). Comparable results
were also obtained after adjusting the model for age, gender, body mass index, glomerular filtration rate,
metabolic syndrome, acute revascularization and ethnicity. A positive correlation was observed between UA
and C reactive protein concentrations in in-hospital deaths only (rho 0.41, p = 0.027).
Conclusion: In patients with acute coronary syndrome, uricemia levels above the current international reference
limit (6.0 mg/dl) were associated with in-hospital mortality, independently from ethnicity and renal function.
© 2017 Published by Elsevier Ireland Ltd.

1. Introduction levels of uric acid predict the development of arterial hypertension [10,
12], obesity [13] and diabetes [14]. However, it is still controversial
Acute coronary syndrome (ACS), a major clinical manifestation of whether uric acid is an independent predictor of cardiovascular disease
atherothrombosis, refers to a wide spectrum of clinical presentations in- [15]. A recent retrospective study has demonstrated that elevated levels
cluding ST-segment elevation myocardial infarction (STEMI), non-ST- of uric acid are an independent predictor of 1-year mortality in patients
segment elevation myocardial infarction (NSTEMI) and unstable angina with ACS treated with percutaneous coronary intervention in a single
[1]. Emerging evidence indicates that plaque phenotype, coronary blood site [16].
flow, endothelial dysfunction, microvascular dysfunction and inflam- The aim of this study was to investigate the prognostic role of serum
mation are pivotal mechanisms contributing to the onset of ACS [2]. uric acid (UA) on admission during a middle term follow up of 180 days
Uric acid seems to be involved in many of these processes, thus contrib- after hospital admission in a multi-ethnic international cohort of pa-
uting to atherosclerosis, plaque composition and vascular instability [3– tients with the entire spectrum of ACS.
6]. Furthermore, uric acid may be an independent risk factor for both
cardiovascular disease [7–10] and kidney disease [10–11] and elevated 2. Material and methods

2.1. Study design


⁎ Corresponding author at: IRCCS Ospedale San Raffaele and Università Vita-Salute San
Raffaele, Via Olgettina 58, 20132 Milan, Italy. This retrospective analysis included patients with ACS who participated at the
E-mail address: cianflone.domenico@hsr.it (D. Cianflone). Stratificazione Prognostica Angina Instabile (SPAI) [17] and First Acute Myocardial Infarc-
1
This author takes responsibility for all aspects of the reliability and freedom from bias tion (FAMI) [18] studies with available blood samples collected on admission, and appro-
of the data presented and their discussed interpretation. priately stored for the measurement of UA concentration. The SPAI study was conducted

http://dx.doi.org/10.1016/j.ijcard.2017.04.027
0167-5273/© 2017 Published by Elsevier Ireland Ltd.
26 M. Magnoni et al. / International Journal of Cardiology 240 (2017) 25–29

from December 1997 to December 2001 and included patients with Braunwald class IIIB All statistical analysis and graphics were produced with JMP soft-
unstable angina diagnosed in presence of: (1) ischaemic ECG changes during recurrent
ware (version 11.0.0, SAS Institute Inc., Cary, North Carolina, USA).
chest pain; (2) evidence of myocardial ischaemia during exercise ECG stress test or during
exercise radionuclide studies or pharmacological echocardiographic stress tests (with ei-
ther dipyridamole or dobutamine); and (3) documentation of obstructive (N 50%) stenosis 4. Results
in at least one major epicardial artery during coronary angiography [17]. Patients were
subsequently defined as NSTEMI and unstable angina according to high-sensitivity tropo- A total of 1548 patients with ACS from SPAI and FAMI studies were
nin levels. The FAMI study was conducted from October 2002 to April 2007 and involved
patients who had electrocardiographic evidence of STEMI, no previous history of coronary
included in the analysis. The UA concentrations ranged from 1.4 to
artery disease and reported symptom onset of b6 h [18]. 11.9 mg/dl and the population was divided in 4 quartiles according to
Patients who received allopurinol therapy, those with LVEF b 30% (STEMI) and LVEF the 25th percentile (4.2 mg/dl), median (5.1 mg/dl), and 75th percentile
b 40% (NSTEMI) and undergoing urgent CABG were excluded from the current analysis. (6.0 mg/dl).
Patients were followed by phone interview or clinical examination after 3 and 6 months
Baseline characteristics of each quartile group are listed in Table 1.
from discharge to evaluate clinical outcomes and therapies. Information about death
was obtained from hospital records, death certificates, or phone contact with relatives of Supplementary Table 1 reports also pharmacological treatment at
the patient or the referring physician. Written informed consent was obtained from all pa- discharged. The proportion of patients with NSTEMI/UA and STEMI
tients. The studies were carried out in accordance with the Declaration of Helsinki and was was similar among groups; de novo ACS and premature ACS also oc-
approved by the institutional ethics committee (SPAI Study was approved by Ethics Com- curred with similar frequency. No statistically significant differences
mittee of Università Cattolica del Sacro Cuore, Rome; approval date 22/09/1997; FAMI
Study was approved by Ethics Committee of Ospedale San Raffaele, Milan; approval date
were observed in UA distribution between European and Chinese pa-
8/11/2001). tients (Table 1).
The prevalence of men, patients with arterial hypertension and met-
abolic syndrome tended to be significantly higher with the increasing
2.2. Clinical evaluation
quartiles of UA. BMI, Framingham Risk Score, triglycerides and serum
Each patient enrolled in the SPAI and FAMI studies underwent a physical examina-
creatinine showed a graded increase, whereas HDL-cholesterol and
tions and standard ECG. Reference blood pressure was measured during the stable eGFR decreased according to the UA quartile.
phase, after recovery from the acute phase or immediately before discharge. Information In-hospital deaths and MACEs were assessed in the overall popula-
about demographic factors, socioeconomic factors (income, employment, level of educa- tion and patients suffering from de novo ACS. In both settings, a higher
tion), lifestyle (physical activity during leisure and work time, dietary habits), psychoso-
proportion of in-hospital deaths was observed in quartile 4 while
cial factors, and personal and family history of cardiovascular disease and traditional risk
factors (smoking, diabetes, dyslipidaemia, hypertension) was collected in a structured MACEs frequencies were similar across groups (Table 2).
standardized questionnaire [17,18]. Height, weight, and abdominal circumference were Considering UA as a categorical variable (cut off: 6.0 mg/dl, quartile
determined. Previous history of CHD, peripheral vascular disease and chronic renal disease 4), UA N6.0 mg/dL increased the risk of in-hospital mortality in both
were also recorded.
overall population (OR 2.9, 95% CI 1.4–6.1, p = 0.0057) and de novo
Non-fasting blood samples were obtained immediately upon admission. Blood was
centrifuged, and serum and plasma samples were bar-coded and frozen at −80 °C until
ACS (OR 3.2, 95%CI 1.5–6.8, p = 0.0033). Multivariate logistic regression
assayed at a core laboratory. Lipids were measured with standard, automated laboratory analysis showed that the adjusted model for age, gender, eGFR classes,
methods and serum C reactive protein was assayed by a high-sensitivity nephelometric and metabolic syndrome (Model 1), obtained similar results (Overall
method (Nephelometric 100 Analyzer; Behring, Scoppito, Italy). population: OR 2.8, 95% CI 1.2–6.5, p = 0.0143; de novo ACS: OR 3.1,
In the present study, for each patient with available blood samples stored in a dedicat-
95% CI 1.3–7.4, p = 0.0095). In addition to the variables considered in
ed biological bank, serum creatinine and serum uric (UA) acid were measured in a central
laboratory (Clinical Laboratory Service, IRCCS Ospedale San Raffaele, Milano), in a single the Model 1, two further variables were evaluated: revascularization
batch, by personnel unaware of patients' characteristics. UA concentrations were deter- (Model 2) and revascularization and ethnicity (Model 3). In Model 2, in
mined with standard, automated laboratory methods with an enzymatic colorimetric the overall population OR was 2.8 (95% CI 1.2–6.4, p = 0.0187), while in
test on a Cobas C 6000 (Roche Diagnostics, Monza Italy); the coefficient of variation was patients with de novo ACS OR was 3.0 (95% CI 1.3–7.4, p = 0.016); in
2%, measured. Renal function was assessed through serum creatinine determination and
estimated glomerular filtration rate (eGFR) was obtained using the four-component Mod-
Model 3, in overall population OR was 2.7 (95%CI 1.2–6.3, p = 0.0226)
ified Diet Renal Disease (MDRD) formula. and in patients with de novo ACS OR was 3.0 (95%CI 1.3–7.3, p =
0.0127) (Table 3).
Furthermore, the correlation between UA and C-reactive protein
3. Statistical analysis (CRP) levels was assessed in patients who survived at the follow up, in
patients who died during hospitalization and after hospital discharge.
Continuous variables were reported as mean and standard deviation UA and CRP concentrations positively correlated in in-hospital dead pa-
or median and interquartile range according to their distribution; com- tients (rho 0.42, p = 0.038) only, whilst the correlation between UA and
parison between groups were performed with ANOVA test and 2-sided CRP concentration was not observed in both survived patients and in
Student's t-test or Wilcoxon test and Kruskal-Wallis test, as appropriate. patients who died after discharge (Fig. 1).
Categorical variables were reported as percentage and compared with
chi-square test. A p value b 0.05 was considered statistically significant. 5. Discussion
Univariate correlation was obtained with a Spearman Rank-Order
Correlation: Multivariate logistic regression models were used to assess The present study describes the relationship among UA, cardiovas-
the relationship between UA and outcomes. Estimates of odds ratios cular risk factors and outcome in a wide cohort of patients with ACS.
with their 95% confidence intervals were reported. First, UA was significantly associated with other well-established risk
The study population was divided in four groups, per UA concentra- factors for cardiovascular disease, namely arterial hypertension, BMI,
tion. UA quartiles were: quartile 1 ≤ 4.2 mg/dl; quartile 2: 4.3 to HDL-cholesterol, triglycerides, the main component metabolic syn-
5.1 mg/dl; quartile 3: 5.2 to 6.0 and quartile 4 N 6.0 mg/dl. The primary drome and renal function. The role of UA as independent cardiovascular
endpoint was mortality at 180 days from hospital admission; the num- risk factor is still controversial. For decades, it has been described that
ber of major adverse cardiac events (MACEs) occurring within 180 days uric acid was implicated in cardiovascular disease [7–10,15,19,20].
was assessed; MACEs included death, heart failure, non-fatal stroke, However, even if UA per se might not represent a modifiable direct
non-fatal STEMI. Recurrence of acute coronary syndrome (both STEMI risk factor for cardiovascular disease, it may be considered a predictive
and NSTEMI) was also evaluated. marker for cardiovascular risk that can worsen other established risk
Premature ACS was defined as ACS in men aged ≤ 55 years and factors. In fact, evidence indicates that uric acid plays a role in hyperten-
women aged ≤ 65 years, following the criteria of premature ischemic sion, obesity, and diabetes [10–14].
heart disease as applied to the definition of the family history for ische- Second, our study showed that elevated UA concentrations
mic heart disease. (N6.0 mg/dl) on admission were associated with an increased risk of
M. Magnoni et al. / International Journal of Cardiology 240 (2017) 25–29 27

Table 1
Baseline characteristics by quartiles of Serum Uric Acid (UA).

UA quartile (mg/dl)

1 2 3 4 p value

n = 386 n = 420 n = 359 n = 383


Demographic
Age, mean (SD), yrs 63.0 ± 11.2 63.4 ± 11.2 63.0 ± 11.4 61.4 ± 12.5 0.085
Males, no. (%) 231 (59.8) 294 (70.0) 281 (78.3) 315 (82.3) b0.0001
Ethnicity 0.52
Caucasian, no. (%) 288 (74.6) 313 (74.5) 258 (72.0) 271 (70.8)
Chinese, no. (%) 98 (25.4) 107 (25.5) 101 (28.0) 112 (29.2)
Clinical presentation 0.62
STEMI, no. (%) 231 (59.8) 263 (62.6) 219 (61.0) 246 (64.2)
NSTEMI/UA, no. (%) 155 (40.2) 157 (37.4) 140 (39.0) 137 (35.8)
De novo ACS, no. (%) 300 (77.7) 328 (78.1) 275 (76.6) 294 (76.8) 0.95
Premature ACS, no. (%) 145 (37.6) 139 (33.1) 116 (32.3) 140 (36.6) 0.34
CV risk factors
Family history of IHD, no. (%) 98 (25.4) 112 (26.7) 91 (25.4) 89 (23.2) 0.73
Arterial hypertension, no. (%) 164 (42.5) 212 (50.5) 167 (46.5) 216 (56.4) 0.001
Diabetes mellitus, no. (%) 74 (19.2) 84 (20.0) 50 (13.9) 67 (17.5) 0.13
Current smoking, no. (%) 83 (21.5) 105 (25.0) 69 (19.2) 78 (20.4) 0.57
BMI, mean (SD), kg/m2 25.7 ± 3.9 26.0 ± 3.7 26.5 ± 3.6 27.0 ± 3.9 b0.0001
Framingham risk score, mean (SD), % 13.6 ± 7.9 15.9 ± 7.9 16.2 ± 7.9 17.4 ± 7.7 b0.0001
Metabolic syndrome, no. (%) 108 (28) 150 (35.7) 136 (37.9) 172 (44.9) b0.0001
Laboratory data
Total cholesterol, mean (SD), mg/dl 212.6 ± 44.5 216.9 ± 48.3 214.8 ± 53.5 220.6 ± 49.4 0.15
LDL-Cholesterol, mean (SD), mg/dl 143.9 ± 45.2 150.0 ± 47.6 147.6 ± 55.8 150.7 ± 49.6 0.30
HDL-Cholesterol, mean (SD), mg/dl 44.2 ± 12.8 42.4 ± 11.7 41.1 ± 10.1 40.2 ± 10.5 b0.0001
Triglycerides, median (IQR), mg/dl 114.7 129.5 131.8 150.8 b0.0001
(78.8–158.4) (92.1–175.1) (95.0–183.3) (110.3–209.0)
C-Reactive protein, median (IQR), mg/l 2.7 2.8 3.4 3.3 0.20
(1.4–6.8) (1.3–6.3) (1.4–7.2) (1.6–7.5)
Serum creatinine, mean (SD), mg/dl 0.85 ± 0.22 0.97 ± 0.44 1.05 ± 0.53 1.23 ± 1.09 b0.0001
eGFR median (IQR), ml/min/173m2 96.6 85.3 78.4 70.0 b0.0001
(74.9–120.7) (66.2–108.9) (61.1–99.6) (51.3–90.5)

STEMI, ST-segment elevation myocardial infarction; NSTEMI, non-ST segment elevation myocardial infarction; UA, unstable angina; ACS, acute coronary syndrome; CV, cardiovascular;
IHD, ischemic heart disease; BMI, body mass index; SD, standard deviation; LDL, Low density lipoprotein; HDL, high density lipoprotein; IQR, interquartile range; eGFR, enhanced glomer-
ular filtration rate.

in-hospital mortality independently from age, gender, renal function, with STEMI, NSTEMI and unstable angina, Ndrepepa et al. showed that
metabolic syndrome, revascularization procedure and ethnicity. Con- every 1 mg/dl increase in UA corresponded to the adjusted risk for 1-
versely, no association was observed with MACEs, 180 days-mortality, year mortality increase by 12% [16]. Subgroup analysis in accordance
mortality and non-fatal myocardial infarction and recurrence of ACS. with different clinical presentations of the entire spectrum of ACS
It has been previously reported that in STEMI patients, who showed that in presence of a concentration of UA N 7.5 mg/dl the risk
underwent percutaneous coronary intervention, higher UA levels for 1-year mortality was significantly increased in all types of ACS.
(N6.5 mg/dl) were detectable in the 21.5% and were independently as- In a Japanese cohort of 1124 patients with acute myocardial infarc-
sociated with in-hospital mortality [21]. The same group observed, in a tion (MI), similar levels of UA (N7.5 mg/dl) resulted the best cut-off to
larger STEMI population, the association between higher UA level and predict 30 days and long-term mortality, independently of Killip class
in-hospital complication but the association with mortality was lost [23].
after adjustment for renal function and myocardial damage [22]. In an- Our findings demonstrated the prognostic value of UA on mortality
other single-site retrospective analysis including a large population in patients with ACS, but it was limited to short-term outcome only.

Table 2
Outcome distribution by quartiles of Serum Uric Acid (UA) in overall population and in patients with de novo ACS.

UA quartile (mg/dl)

Overall 1 2 3 4 p value

Overall
In-hospital death. no. (%) 29 (1.9) 5 (1.3) 6 (1.4) 4 (1.1) 14 (3.7) 0.017
180 days outcome (n 1431)
MACE no. (%) 199 (13.9) 43 (12.0) 57 (14.6) 45 (13.8) 54 (15.2) 0.62
Death no. (%) 69 (4.8) 16 (4.5) 19 (4.9) 12 (3.7) 22 (6.2) 0.47
Death/MI no. (%) 94 (6.6) 18 (5.0) 27 (6.9) 22 (6.7) 27 (7.6) 0.55
Recurrent ACS no. (%) 125 (8.7) 27 (7.5) 38 (9.7) 32 (9.8) 28 (7.9) 0.88
De novo ACS
In-hospital death no. (%) 28 (2.3) 5 (1.7) 6 (1.8) 3 (1.1) 14 (4.8) 0.015
180 days outcome (n 1088)
MACE no. (%) 119 (10.9) 29 (10.6) 33 (11.1) 21 (8.6) 36 (13.3) 0.38
Death no. (%) 47 (4.3) 11 (4.0) 11 (3.7) 7 (2.9) 18 (6.7) 0.16
Death/MI no. (%) 61 (5.6) 11 (4.0) 17 (5.7) 12 (4.9) 21 (7.8) 0.26
Recurrent ACS no. (%) 61 (5.6) 16 (5.8) 18 (6.0) 13 (5.3) 14 (5.2) 0.67

UA, serum uric acid; MACE, major adverse cardiovascular event; ACS, acute coronary syndrome; MI, myocardial infarction.
28 M. Magnoni et al. / International Journal of Cardiology 240 (2017) 25–29

Table 3
Logistic regression models for the association between UA and in-hospital mortality in overall population and in patients with de novo ACS.

Unadjusted Model 1 Model 2 Model 3

Overall OR (95%CI) p value OR (95%CI) p value OR (95%CI) p value OR (95%CI) p value


UA N 6.0 vs ≤6.0 mg/dl 2.9 (1.4–6.1) 0.0057 2.8 (1.2–6.5) 0.0143 2.8 (1.2–6.4) 0.0187 2.7 (1.2–6.3) 0.0226
De novo ACS OR (95%CI) p value OR (95%CI) p value OR (95%CI) p value OR (95%CI) p value
UA N 6.0 vs ≤6.0 mg/dl 3.2 (1.5–6.8) 0.0033 3.1 (1.3–7.4) 0.0095 3.0 (1.3–7.4) 0.0106 3.0 (1.3–7.3) 0.0127

Model 1: adjusted for age, gender, eGFR (categorical classes ≥90, 89–60 and b60 ml/min/173 m2), metabolic syndrome.
Model 2: adjusted for age, gender, eGFR, metabolic syndrome, revascularization.
Model 3: adjusted for age, gender, eGFR, metabolic syndrome, revascularization, ethnicity.

As compared to the results of previous studies, a younger age, lower UA Previous studies reported that UA and inflammation markers were
threshold for the upper quartile (6.0 mg/dl) and a lower incidence of positively correlated in apparently healthy individuals [5,32,33], in pa-
short- and mid-term mortality rates probably limited the ability of UA tients with ACS [16] and in patients with chronic heart failure [34].
to predict the long-term mortality in our study. However, for the same Mechanisms underlying the role of uric acid in inflammation remain
reasons, the predictive value as well as the pathophysiological role of to be clarified. However, it has been observed that uric acid induced
UA in the setting of ACS is strengthened. Furthermore, unlike previous the release of MCP-1, IL-1β, IL-6, and TNFα [35,36]; in vitro, it promoted
studies, elevated UA levels and in-hospital mortality risk were signifi- vascular smooth muscle cells, favoured pro-inflammatory response, and
cantly associated in patients with ACS as the first clinical manifestation induced Cox-2 up-regulation and endothelial CRP production [36]. Pro-
of ischemic heart disease (de novo ACS), thus supporting the need to in- inflammatory cytokines (TNFα, IL-1, INFγ), ROS-induced cell damages
clude UA in primary preventive strategies. and apoptosis activated xanthine oxidase to synthesize uric acid,
On the other hand, as recently demonstrated by the GISSI- which may promote pro-inflammatory responses, independently from
Prevenzione Trial, higher UA levels measured within 3 months of MI in its plasmatic concentration [37].
survived patients predicted long-term mortality, thus highlighting the In this context, we found a positive relationship between UA and C-
importance of considering UA as biomarker also for secondary prevention reactive protein concentrations only in patients who died during the
[24]. hospitalization, whereas in the whole study population and in patients
Pathophysiological mechanisms involved in the increased in- who survived or died after the discharge a significant correlation was
hospital mortality in patients with elevated UA levels on admission not observed, likely due to the widespread and multifactorial inflamma-
are not completely understood. Increased UA levels enable to identify tion during ACS. These findings support the hypothesis that a strictly re-
a subgroup more prone to in-hospital adverse outcome, probably be- lationship between oxidative stress and inflammation during ACS may
cause UA reflects several complex adverse mechanisms ranging from promote enhanced pathological disorders, leading to adverse outcomes.
pre-existing relationship with other risk factors to the degree of myo- The study had some limitations. The follow-up duration did not
cardial ischemia and the relative acute metabolic and inflammatory re- allow to assess a long-term association between UA levels and mortali-
sponse. Among cardiovascular risk factors, renal dysfunction is strictly ty. Long-term studies on ACS patients are advisable to better define a
related to cardiovascular mortality and coronary disease. Kowalczyk prognostic value of UA, also within subclinical values, in patients with
et al. reported that UA concentration predicted short- and long-term ACS and concomitant cardiovascular risk factors and further clarify the
mortality in patients with renal dysfunction who experienced MI and potential role of uric acid as cardiovascular risk factor. Although patients
were treated with PCI [25]. In our study, the predictive value of UA of were prospectively recruited in the original studies [17,18], the analysis
in-hospital mortality resulted independent from renal function classes. was retrospective.
Reflecting xanthine oxidase activity, UA is considered a marker of in- More than a quarter of patients included in the analysis were Chi-
creased oxidative stress and, thus, linked with several pathological pro- nese; therefore, our results may be independent from genetic co-
cesses that may promote the worsening of adverse conditions during founders which may be present in more restricted or national trials
myocardial ischemia, such as LDL-cholesterol oxidation and lipid perox- [16,23]. In addition, to obtain a more representative picture of the
idation processes [26–27], renin–angiotensin system [28], platelet [29] real-world practice, we did not restrict the selection of patients who
and inflammatory activation [5]. Moreover, elevated UA levels contrib- only underwent PCI, as previously done [16,22], and included the entire
ute to impaired myocardial blood flow through more severe coronary spectrum of ACS ranging from myocardial infarction to unstable angina.
microvascular dysfunction [30,31].

Fig. 1. Correlation between UA and CRP concentration.


M. Magnoni et al. / International Journal of Cardiology 240 (2017) 25–29 29

Supplementary data to this article can be found online at http://dx. Maseri, FAMI Study Investigators, High-sensitivity C-reactive protein is within nor-
mal levels at the very onset of first ST-segment elevation acute myocardial infarc-
doi.org/10.1016/j.ijcard.2017.04.027. tion in 41% of cases: a multiethnic case-control study, J. Am. Coll. Cardiol. 58
(2011) 2654–2661.
Disclosure [19] P. Verdecchia, G. Schillaci, G.P. Reboldi, F. Sariteu-Sanio, C. Porcellati, P. Brunetti, Re-
lation between serum uric acid and risk of cardiovascular disease in essential hyper-
tension. The PIUMA study, Hyperfetisioit 36 (2000) 1072–1078.
The authors report no relationships that could be construed as a con- [20] G. Savarese, C. Ferri, B. Trimarco, G. Rosano, S. Dellegrottaglie, T. Losco, L. Casaretti, C.
flict of interest. D'Amore, F. Gambardella, M. Prastaro, G. Rengo, D. Leosco, P. Perrone-Filardi, Chang-
es in serum uric acid levels and cardiovascular events: a meta-analysis, Nutr. Metab.
Cardiovasc. Dis. 23 (2013) 707–714.
Acknowledgement [21] C. Lazzeri, S. Valente, M. Chiostri, A. Sori, P. Bernardo, G.F. Gensini, Uric acid in the
acute phase of ST elevation myocardial infarction submitted to primary PCI: its
prognostic role and relation with inflammatory markers: a single center experience,
We thank Elisa Sala, PhD, Independent Medical Writer, for her med-
Int. J. Cardiol. 138 (2010) 206–209.
ical editorial assistance with our report. We thank Mosè Barbaro and [22] C. Lazzeri, S. Valente, M. Chiostri, C. Picariello, G.F. Gensini, Uric acid in the early risk
Michele Raso for the technical assistance in uric acid measurement. stratification of ST elevation myocardial infarction, Intern. Emerg. Med. 7 (2012)
We thank Menarini International Operations Luxembourg S.A. for their 33–39.
[23] S. Kojima, T. Sakamoto, M. Ishihara, K. Kimura, S. Miyazaki, M. Yamagishi, C. Tei, H.
partial financial support by an unrestricted grant. Hiraoka, M. Sonoda, K. Tsuchihashi, N. Shimoyama, T. Honda, Y. Ogata, K. Matsui,
H. Ogawa, Japanese Acute Coronary Syndrome Study (JACSS) Investigators, Prog-
References nostic usefulness of serum uric acid after acute myocardial infarction (the
Japanese acute coronary syndrome study), Am. J. Cardiol. 96 (2005) 489–495.
[1] P. Libby, Mechanisms of acute coronary syndromes and their implications for ther- [24] G. Levantesi, R.M. Marfisi, M.G. Franzosi, A.P. Maggioni, G.L. Nicolosi, C. Schweiger,
apy, N. Engl. J. Med. 368 (2013) 2004–2013. M.G. Silletta, L. Tavazzi, G. Tognoni, R. Marchioli, Uric acid: a cardiovascular risk fac-
[2] D.C. Tong, A.M. Wilson, J. Layland, Novel risk factors for acute coronary syndromes tor in patients with recent myocardial infarction, Int. J. Cardiol. 167 (1) (2013)
and emerging therapies, Int. J. Cardiol. 220 (2016) 815–824. 262–269.
[3] R. Rodrigo, M. Libuy, F. Feliú, D. Hasson, Oxidative stress-related biomarkers in es- [25] J. Kowalczyk, P. Francuz, R. Swoboda, R. Lenarczyk, B. Sredniawa, A. Golda, T. Kurek,
sential hypertension and ischemia-reperfusion myocardial damage, Dis. Markers M. Mazurek, T. Podolecki, L. Polonski, Z. Kalarus, Prognostic significance of hyperuri-
35 (2013) 773–790. cemia in patients with different types of renal dysfunction and acute myocardial in-
[4] U.M. Khosla, S. Zharikov, J.L. Finch, T. Nakagawa, C. Roncal, W. Mu, K. Krotova, E.R. farction treated with percutaneous coronary intervention, Nephron Clin. Pract. 116
Block, S. Prabhakar, R.J. Johnson, Hyperuricemia induces endothelial dysfunction, (2) (2010) c114–c122.
Kidney Int. 67 (2005) 1739–1742. [26] A.F. Cicero, M. Rosticci, M. Cagnati, R. Urso, G. Scapagnini, M. Morbini, E. Grandi, S.
[5] C. Ruggiero, A. Cherubini, A. Ble, A.J. Bos, M. Maggio, V.D. Dixit, F. Lauretani, S. D'Addato, C. Borghi, Brisighella Heart Study Group, Serum uric acid and markers
Bandinelli, U. Senin, L. Ferrucci, Uric acid and inflammatory markers, Eur. Heart J. of low-density lipoprotein oxidation in nonsmoking healthy subjects: data from
27 (2006) 1174–1181. the Brisighella heart study, Pol. Arch. Med. Wewn. 124 (12) (2014) 661-8.
[6] K. Ando, H. Takahashi, T. Watanabe, H. Daidoji, Y. Otaki, S. Nishiyama, T. Arimoto, T. [27] I.K. De Scheerder, A.M. van de Kraay, J.M. Lamers, J.F. Koster, J.W. de Jong, P.W.
Shishido, T. Miyashita, T. Miyamoto, I. Kubota, Impact of serum uric acid levels on Serruys, Myocardial malondialdehyde and uric acid release after short-lasting coro-
coronary plaque stability evaluated using integrated backscatter intravascular ultra- nary occlusions during coronary angioplasty: potential mechanisms for free radical
sound in patients with coronary artery disease, J. Atheroscler. Thromb. 23 (8) (Aug 1 generation, Am. J. Cardiol. 68 (1991) 392–395.
2016) 932-9. [28] D.B. Corry, P. Eslami, K. Yamamoto, M.D. Nyby, H. Makino, M.L. Tuck, Uric acid stim-
[7] J. Fang, M.H. Alderman, Serum uric acid and cardiovascular mortality the NHANES I ulates vascular smooth muscle cell proliferation and oxidative stress via the vascular
epidemiologic follow-up study, 1971–1992. National Health and Nutrition Examina- renin-angiotensin system, J. Hypertens. 26 (2008) 269-75.
tion survey, JAMA 283 (2000) 2404–2410. [29] M. Alderman, K.J. Aiyer, Uric acid: role in cardiovascular disease and effects of
[8] M.H. Alderman, H. Cohen, S. Madhavan, S. Kivlighn, Serum uric acid and cardiovas- losartan, Curr. Med. Res. Opin. 20 (2004) 369–379.
cular events in successfully treated hypertensive patients, Hypertension 34 (1999) [30] M. Akpek, M.G. Kaya, H. Uyarel, M. Yarlioglues, N. Kalay, O. Gunebakmaz, O. Dogdu, I.
144–150. Ardic, D. Elcik, O. Sahin, A. Oguzhan, A. Ergin, C.M. Gibson, The association of serum
[9] M.J. Bos, P.J. Koudstaal, A. Hofman, J.C. Witteman, M.M. Breteler, Uric acid is a risk uric acid levels on coronary flow in patients with STEMI undergoing primary PCI,
factor for myocardial infarction and stroke: the Rotterdam study, Stroke 37 (2006) Atherosclerosis 219 (2011) 334e41.
1503e7. [31] N. Basar, N. Sen, F. Ozcan, G. Erden, S. Kanat, E. Sokmen, A. Isleyen, H. Yuzgecer, M.F.
[10] C. Borghi, E.A. Rosei, T. Bardin, J. Dawson, A. Dominiczak, J.T. Kielstein, A.J. Manolis, F. Ozlu, M. Yildirimkaya, O. Maden, A. Covic, R.J. Johnson, M. Kanbay, Elevated serum
Perez-Ruiz, G. Mancia, Serum uric acid and the risk of cardiovascular and renal dis- uric acid predicts angiographic impaired reperfusion and 1-year mortality in ST-
ease, J. Hypertens. 33 (9) (2015) 1729-41. segment elevation myocardial infarction patients undergoing percutaneous coro-
[11] K. Iseki, Y. Ikemiya, T. Inoue, C. Iseki, K. Kinjo, S. Takishita, Significance of hyperuri- nary intervention, J. Investig. Med. 59 (2011) 931e7.
cemia as a risk factor for developing ESRD in a screened cohort, Am. J. Kidney Dis. 44 [32] C. Ruggiero, A. Cherubini, E. Miller III, M. Maggio, S.S. Najjar, F. Lauretani, S.
(2004) 642–650. Bandinelli, U. Senin, L. Ferrucci, Usefulness of uric acid to predict changes in C-
[12] A. Shankar, R. Klein, B.E. Klein, F.J. Nieto, The association between serum uric acid reactive protein and interleukin-6 in 3-year period in Italians aged 21 to 98 years,
level and long-term incidence of hypertension: population-based cohort study, J. Am. J. Cardiol. 100 (2007) 115–121.
Hum. Hypertens. 20 (2006) 937–945. [33] M. Fröhlich, A. Imhof, G. Berg, W.L. Hutchinson, M.B. Pepys, H. Boeing, R. Muche, H.
[13] K. Masuo, H. Kawaguchi, H. Mikami, T. Ogihara, M.L. Tuck, Serum uric acid and plas- Brenner, W. Koenig, Association between C-reactive protein and features of the met-
ma norepinephrine concentrations predict subsequent weight gain and blood pres- abolic syndrome: a population-based study, Diabetes Care 23 (2000) 1835-9.
sure elevation, Hypertension 42 (2003) 474–480. [34] F. Leyva, S.D. Anker, I.F. Godsland, M. Teixeira, P.G. Hellewell, W.J. Kox, P.A. Poole-
[14] A. Dehghan, M. van Hoek, E.J. Sijbrands, A. Hofman, J.C. Witteman, High serum uric Wilson, A.J. Coats, Uric acid in chronic heart failure: a marker of chronic inflamma-
acid as a novel risk factor for type 2 diabetes, Diabetes Care 31 (2008) 361–362. tion, Eur. Heart J. 19 (1998) 1814–1822.
[15] B.F. Culleton, M.G. Larson, W.B. Kannel, D. Levy, Serum uric acid and risk for cardio- [35] J. Kanellis, S. Watanabe, J.H. Li, D.H. Kang, P. Li, T. Nakagawa, A. Wamsley, D. Sheikh-
vascular disease and death: the Framingham heart study, Ann. Intern. Med. 131 Hamad, H.Y. Lan, L. Feng, R.J. Johnson, Uric acid stimulates monocyte
(1999) 7–13. chemoattractant protein-1 production in vascular smooth muscle cells via
[16] G. Ndrepepa, S. Braun, H.U. Haase, S. Schulz, S. Ranftl, M. Hadamitzky, J. Mehilli, A. mitogen-activated protein kinase and cyclooxygenase-2, Hypertension 416 (2003)
Schömig, A. Kastrati, Prognostic value of uric acid in patients with acute coronary 1287–1293.
syndromes, Am. J. Cardiol. 109 (2012) 1260–1265. [36] R.J. Johnson, B. Rodriguez-Iturbe, D.H. Kang, D.I. Feig, J. Herrera-Acosta, A unifying
[17] G.A. Lanza, D. Cianflone, A.G. Rebuzzi, G. Angeloni, A. Sestito, G. Ciriello, G. La Torre, pathway for essential hypertension, Am. J. Hypertens. 18 (2005) 431–440.
F. Crea, A. Maseri, Stratificazione Prognostica dell'Angina Instabile Study [37] Y. Shi, J.E. Evans, K.L. Rock, Molecular identification of a danger signal that alerts the
Investigators, Prognostic value of ventricular arrhythmias and heart rate variability immune system to dying cells, Nature 425 (2003) 516–521.
in patients with unstable angina, Heart 92 (2006) 1055–1063.
[18] N. Cristell, D. Cianflone, A. Durante, E. Ammirati, D. Vanuzzo, M. Banfi, G. Calori, A.
Latib, F. Crea, G. Marenzi, M. De Metrio, L. Moretti, H. Li, N.G. Uren, D. Hu, A.

You might also like