You are on page 1of 27

AHA Scientific Statement

Pharmacotherapy in Chronic Kidney Disease Patients


Presenting With Acute Coronary Syndrome
A Scientific Statement From the American Heart Association
Jeffrey B. Washam, PharmD, FAHA, Chair; Charles A. Herzog, MD, FAHA;
Amber L. Beitelshees, PharmD, MPH, FAHA; Mauricio G. Cohen, MD;
Timothy D. Henry, MD; Navin K. Kapur, MD; Jessica L. Mega, MD, MPH, FAHA;
Venu Menon, MD, FAHA; Robert L. Page II, PharmD, MSPH, FAHA;
L. Kristin Newby, MD, MHS, FAHA, Co-Chair; on behalf of the American Heart Association Clinical
Pharmacology Committee of the Council on Clinical Cardiology, Council on Cardiovascular Surgery
and Anesthesia, Council on Functional Genomics and Translational Biology, Council on the Kidney in
Cardiovascular Disease, and Council on Quality of Care and Outcomes Research

C hronic kidney disease (CKD) is frequently encoun-


tered among patients presenting with acute coro-
nary syndrome (ACS). Recent data from the National
Background and CKD Staging
It has been appreciated now for more than a decade that CKD
is a powerful independent predictor of cardiovascular mor-
Cardiovascular Data Registry–Acute Coronary Treatment and bidity, cardiovascular mortality, and all-cause mortality. The
Intervention Outcomes Network (NCDR-ACTION) reported systematic classification of CKD in large part is based on the
CKD (defined as estimated creatinine clearance [CrCl] efforts of Andrew Levey and colleagues, who published the
<60 mL·min−1·1.73 m−2) prevalence rates of 30.5% among K/DOQI (Kidney Disease Outcomes Quality Initiative) clini-
patients presenting with ST-segment–elevation myocardial cal practice guidelines for CKD.7 The original schema some-
infarction (STEMI) and 42.9% among patients present- what arbitrarily defined stages 1 to 5 CKD on the basis of
ing with non–ST-segment–elevation myocardial infarction estimated glomerular filtration rate (eGFR) in the following
(NSTEMI).1 The presence of CKD among patients presenting manner: Stage 1, eGFR ≥90 mL·min−1·1.73 m−2 (with evi-
Downloaded from http://ahajournals.org by on January 29, 2019

with ACS has been associated with worse outcomes, including dence of kidney damage present, such as albuminuria); stage
higher rates of mortality and bleeding.2–4 Despite the increased 2, eGFR <90 but ≥60 (with evidence of kidney damage such
risk for adverse outcomes, CKD patients presenting with ACS as albuminuria); stage 3, eGFR <60 but ≥30; stage 4, eGFR
are less likely to receive evidence-based therapies, including <30 but ≥15; and stage 5, eGFR <15 or undergoing dialysis.7
medications.1 In addition, patients with CKD have been under- An additional modification was made to create a stage 3a
represented in randomized controlled trials of ACS pharmaco- (eGFR 45–59 mL·min−1·1.73 m−2) and stage 3b (eGFR 30–44
therapy.5,6 Thus, the net effect is a relative lack of evidence and mL·min−1·1.73 m−2). Most recently, based on the stepwise
potential for uncertainty in selecting medications in this high- association of albuminuria with increased rates of CKD
risk population. The purpose of this scientific statement is to progression, cardiovascular mortality, and total mortality,
provide a comprehensive review of the published literature the Kidney Disease: Improving Global Outcomes (KDIGO)
and provide recommendations on the use of evidence-based group has recommended altering the classification scheme to
pharmacotherapies in CKD patients presenting with ACS. include urinary albumin excretion (Figure 1).8

The American Heart Association makes every effort to avoid any actual or potential conflicts of interest that may arise as a result of an outside relationship
or a personal, professional, or business interest of a member of the writing panel. Specifically, all members of the writing group are required to complete
and submit a Disclosure Questionnaire showing all such relationships that might be perceived as real or potential conflicts of interest.
This statement was approved by the American Heart Association Science Advisory and Coordinating Committee on September 15, 2014. A copy of the
document is available at http://my.americanheart.org/statements by selecting either the “By Topic” link or the “By Publication Date” link. To purchase
additional reprints, call 843-216-2533 or e-mail kelle.ramsay@wolterskluwer.com.
The American Heart Association requests that this document be cited as follows: Washam JB, Herzog CA, Beitelshees AL, Cohen MG, Henry TD,
Kapur NK, Mega JL, Menon V, Page RL 2nd, Newby LK; on behalf of the American Heart Association Clinical Pharmacology Committee of the Council
on Clinical Cardiology, Council on Cardiovascular Surgery and Anesthesia, Council on Functional Genomics and Translational Biology, Council on the
Kidney in Cardiovascular Disease, and Council on Quality of Care and Outcomes Research. Pharmacotherapy in chronic kidney disease patients presenting
with acute coronary syndrome: a scientific statement from the American Heart Association. Circulation. 2015;131:1123–1149.
Expert peer review of AHA Scientific Statements is conducted by the AHA Office of Science Operations. For more on AHA statements and guidelines
development, visit http://my.americanheart.org/statements and select the “Policies and Development” link.
Permissions: Multiple copies, modification, alteration, enhancement, and/or distribution of this document are not permitted without the express
permission of the American Heart Association. Instructions for obtaining permission are located at http://www.heart.org/HEARTORG/General/Copyright-
Permission-Guidelines_UCM_300404_Article.jsp. A link to the “Copyright Permissions Request Form” appears on the right side of the page.
(Circulation. 2015;131:1123-1149. DOI: 10.1161/CIR.0000000000000183.)
© 2015 American Heart Association, Inc.
Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIR.0000000000000183

1123
1124  Circulation  March 24, 2015

Persistent albuminuria categories


Description and range

A1 A2 A3
Prognosis of CKD by GFR
Normal to
and Albuminuria Categories: mildly
Moderately Severely
KDIGO 2012 increased
increased increased Figure 1. Risk for all-cause mortality,
cardiovascular mortality, end-stage
<30 mg/g 30-300 mg/g >300 mg/g
renal disease, progressive chronic
<3 mg/mmol 3-30 mg/mmol >30 mg/mmol kidney disease (CKD), or acute kidney
injury among CKD patients according
to glomerular filtration rate (GFR) and
G1 Normal or high ≥90 albuminuria categories. KDIGO indicates
Kidney Disease: Improving Global
GFR categories (ml/min/ 1.73m2)

Outcomes. Reprinted from Kidney


G2 Mildly decreased 60-89
Disease: Improving Global Outcomes
Description and range

(KDIGO) CKD Work Group8 with


Mildly to moderately permission from Macmillan Publishers
G3a 45-59
decreased
Ltd. Copyright © 2013, International
Moderately to Society of Nephrology.
G3b 30-44
severely decreased

G4 Severely decreased 15-29

G5 Kidney failure <15

As shown in Figure 2, there is a stepwise incremental >2.5 mg/dL), and 274 777 non-CKD patients.14 Those with
age-standardized risk for all-cause mortality, cardiovascu- advanced CKD and dialysis were less likely to have chest
lar events, and hospitalization associated with diminishing pain on admission (40.4% and 41.1%, respectively) than
renal function.9 Compared with patients with eGFR ≥60 those without CKD (61.6%). Similar observations were
mL·min−1·1.73 m−2, the adjusted hazard for death among made in the SWEDEHEART registry [Swedish Web System
patients with eGFR 15 to 29 mL·min−1·1.73 m−2 is more than for Enhancement and Development of Evidence-Based Care
3-fold higher, and nearly 6 times higher for patients with in Heart Disease Evaluated According to Recommended
Downloaded from http://ahajournals.org by on January 29, 2019

eGFR <15 mL·min−1·1.73 m−2. Therapies]; however, up to two thirds of patients with stage
It has been suggested that CKD should be regarded as a 4 and 5 CKD in that registry had chest pain at presentation.15
“coronary heart disease equivalent.” The publication by The USRDS-NRMI study also showed that MI patients with
Tonelli and colleagues,10 using the Alberta Kidney Disease advanced CKD and those undergoing dialysis more often had
Network database and the National Health and Nutrition a diagnosis at presentation other than ACS (44% and 47.7%,
Examination Survey 2003 to 2006, estimated the risk of hos- respectively) compared with patients without CKD (25.8%).
pital admissions for myocardial infarction (MI) and all-cause Compared with patients without CKD, patients with advanced
death among individuals with previous MI, diabetes melli- CKD were also less likely to have ST-segment elevation
tus, or CKD. Among people without previous MI, the risk of (15.9% versus 32.5%, respectively) but more likely to have
MI was lower among patients with diabetes mellitus without heart failure on presentation (52.2% versus 27.2%, respec-
CKD than among those with CKD (5.4 per 1000 patient-years tively) and a higher rate of in-hospital mortality (23% versus
versus 6.9 per 1000 patient-years). The findings in this study 12.6%, respectively).10 Similar differences existed between
would indicate that CKD should be added to the list of criteria those with advanced CKD and those undergoing dialysis.
defining people at high risk for coronary events. The distribution of electrocardiographic presentations varies
according to severity of CKD, with fewer STEMIs and more
Special Clinical Characteristics NSTEMI and left bundle branch block among populations
of ACS in CKD Patients with increasingly worse renal function (Figure 3).11,15
An additional consideration in the diagnosis of ACS in
Clinical Presentation of ACS Among CKD Patients patients with CKD is the interpretation of cardiac biomark-
The clinical presentation of ACS among patients with CKD ers. Chronic troponin elevations in clinically stable patients
is distinctly different from that of patients without CKD in with renal failure have been observed and likely represent
the general population.1,11–15 First, the prevalence of chest nonischemic myocardial injury.16 In spite of these chronic
pain among patients with ACS is inversely related to stage of troponin elevations in a population of patients with CKD,
CKD. As shown in Figure 3, there is a graded reduction in the National Association of Clinical Biochemistry labora-
the frequency of chest pain as eGFR falls.15 For example, in a tory medicine practice guidelines recommend the use of
collaborative project of the United States Renal Data System troponins for the diagnosis of MI in patients with CKD pre-
(USRDS) and the National Registry of Myocardial Infarction senting with symptoms or electrocardiographic changes sug-
(NRMI), the clinical characteristics were compared in a large gestive of myocardial ischemia.17 These guidelines, along
population of MI patients that included 2390 dialysis patients, with other expert writing groups, advise the importance of a
29 319 patients with advanced CKD (serum creatinine [SCr] dynamic change in troponin values after presentation in the
Washam et al   Pharmacotherapy in CKD Patients With ACS   1125

presentation (and thus would not be considered for appropri-


ate therapeutic interventions).

Methods of Estimating Renal


Function for Drug Dosing
Whereas the Modification of Diet in Renal Disease (MDRD)
equation is widely used for CKD diagnosis and staging, the
Cockcroft-Gault (CG) equation has been the most commonly
used equation to estimate renal function for dose adjustment
of medications.22 Although these equations have limitations,
both the CG and MDRD equations have been shown to cor-
relate relatively well with measured glomerular filtration rate
(GFR),22 but differences in medication dose recommendations
have been reported depending on which equation is used.23–25
An analysis of the Can Rapid Risk Stratification of Unstable
Angina Patients Suppress Adverse Outcomes With Early
Implementation of the ACC/AHA Guidelines (CRUSADE)
registry was conducted to compare the CG and MDRD equa-
tions with regard to the recommended doses of eptifibatide,
tirofiban, and enoxaparin.23 Results of this analysis showed
a 20% difference in CKD classification between the 2 equa-
tions. The proportion of patients classified as having normal/
mild CKD (eGFR ≥60 mL/min), moderate CKD (eGFR 30–
59 mL/min), and severe CKD (eGFR <30 mL/min) by the CG
equation was 41.2%, 39.8%, and 19% compared with 58.9%,
31.5%, and 9.6%, respectively, by the MDRD equation. In
addition, marked differences were seen in the proportion of
patients for whom dose adjustment was recommended by
the CG versus MDRD equation, respectively, for eptifibatide
Downloaded from http://ahajournals.org by on January 29, 2019

(45.7% versus 27.3%) and for enoxaparin or tirofiban (19.0%


versus 9.6%). Over the past decade, the CG equation has been
the preferred method used in assessing renal function for dose
adjustment and to determine trial eligibility in randomized
controlled trials of antithrombotic medications. Until further
data validating the MDRD equation as a method for dose
adjustment of cardiovascular medications become available,
current data support the use of the CG equation for cardiovas-
cular drug dosing (Table 1).

Pharmacotherapy for ACS


Figure 2. Age-standardized rates of death of any cause (A),
cardiovascular events (defined as hospitalization for coronary
Among Patients With CKD
heart disease, heart failure, ischemic stroke, or peripheral Fibrinolytic Therapy
arterial disease) (B), and hospitalization (C) according to the
estimated glomerular filtration rate (GFR) among 1 120 295 Current American College of Cardiology Foundation/
ambulatory adults. Reprinted from Go et al9 with permission American Heart Association guidelines give fibrinolytic ther-
from Massachusetts Medical Society. Copyright © 2004, apy a Class I recommendation for STEMI patients present-
Massachusetts Medical Society.
ing within 12 hours of the onset of ischemia symptoms and
without contraindications, when it is anticipated primary per-
identification of acute MI (AMI) in patients with end-stage cutaneous coronary intervention (PCI) cannot be performed
renal disease (ESRD), who more frequently have chronically within 120 minutes.26 Although primary PCI is the preferred
elevated troponin levels.16–18 reperfusion strategy for STEMI patients, recent data from the
This striking difference in clinical presentation and electro- NCDR-ACTION Registry indicate that fibrinolytic therapy
cardiographic findings has implications for correct diagnosis was the initial reperfusion strategy in ≈10% of patients in the
and subsequent treatment. It has been a subject of great atten- United States.29 Because initial randomized controlled trials of
tion that the use of evidence-based therapy is lower among fibrinolytic therapy did not assess the treatment effect of the
patients with CKD.1–3,19–21 Not only are those with ACS and fibrinolysis in the subgroup of patients with CKD, outcome
CKD less likely to receive evidence-based therapies, the data in this population are limited. Clinical trial and observa-
atypical clinical presentation of these patients makes it less tional data on the outcomes of ACS patients with CKD receiv-
likely that they will be correctly identified as having ACS on ing fibrinolytic therapy are summarized in Table 2.
1126  Circulation  March 24, 2015

100%
90%
88%
90% eGFR ≥90 60–89 30–59 15–29 <15/dialysis

80% 76%

67% 66%
70% 65%
60%

Frequency (%)
60%
60% 58%
56%

50% 46%
41%
38% 37%
40%
31% 29%
30% 25%
22%
20% 15% 15%
10% 11% 12%
10% 3%
6%

0%
Chest pain Killip ≥ II STEMI LBBB NSTEMI

Figure 3. Relation of renal function to presentation, symptoms, and ECG changes in patients presenting with acute coronary syndrome.
Data from the SWEDEHEART Registry (Swedish Web System for Enhancement and Development of Evidence-Based Care in Heart
Disease Evaluated According to Recommended Therapies). eGFR indicates estimated glomerular filtration rate; LBBB, left bundle branch
block; NSTEMI, non–ST-segment–elevation myocardial infarction; and STEMI, ST-segment–elevation myocardial infarction. Reprinted
from Szummer et al15 with permission of the publisher. Copyright © 2010, Blackwell Publishing Ltd.

A pooled analysis of 16  710 patients enrolled in the function, and rates of intracranial hemorrhage increased with
Thrombolysis in Myocardial Infarction (TIMI)-10A, TIMI- worsening renal function (0.6%, 0.8%, 1.8%, and 3.0% for
10B, TIMI-14, and Intravenous NPA for the Treatment of normal, mildly impaired, moderately impaired, and severely
Infarcting Myocardium Early (InTIME-II) trials was con- impaired CrCl, respectively; P<0.0001 for trend).
ducted to assess the impact of baseline renal function (SCr and Several observational analyses have evaluated the associa-
CrCl) on outcomes in patients receiving fibrinolytic therapy.30 tion of CKD with outcomes and the treatment effect of fibrino-
A stepwise increase in mortality was seen with worsening renal lytic therapy in STEMI patients with various results. Hobbach
Downloaded from http://ahajournals.org by on January 29, 2019

Table 1.  Doses of Parenteral Antithrombotic Agents


Renal
Medication Elimination Dose in Patients Without CKD Dose Adjustment in CKD
Abciximab26,27 NS • PCI: 0.25-mg/kg bolus followed by infusion No adjustment
of 0.125 μg·kg−1·min−1 (maximum 10 μg/min)
for 12 h after procedure
Bivalirudin26 20% • PCI: 0.75-mg/kg bolus followed by infusion of CrCl <30 mL/min:
1.75 mg·kg−1·h−1 for duration of the procedure • PCI: 0.75-mg/kg bolus followed by infusion of
1 mg·kg−1·h−1 for the duration of the procedure
Dialysis:
• PCI28: 0.75-mg/kg bolus followed by infusion of
0.25 mg·kg−1·h−1
Enoxaparin26,27 40% •UA/NSTEMI: 1 mg/kg SC every 12 h CrCl <30 mL/min:
• STEMI patients <75 y of age receiving fibrinolytic • UA/NSTEMI: 1 mg/kg SC once daily
therapy: 30-mg single IV bolus plus a 1-mg/kg • STEMI patients <75 y of age receiving fibrinolytic therapy:
SC dose followed by 1 mg/kg SC every 12 h 30-mg single IV bolus plus a 1-mg/kg SC dose followed by
• STEMI patients ≥75 y of age receiving fibrinolytic 1 mg/kg administered SC once daily
therapy: No bolus, 0.75 mg/kg SC every 12 h • STEMI patients ≥75 y of age receiving fibrinolytic therapy:
No bolus, 1 mg/kg administered SC once daily
Not recommended in dialysis patients
Eptifibatide7,26 50% • ACS: 180-μg/kg bolus followed by an infusion CrCl < 50 mL/min:
of 2 μg·kg−1·min−1 for up to 72 h • ACS: 180-μg/kg bolus followed by infusion of
• PCI: 180 μg/kg followed by continuous infusion 1 μg·kg−1·min−1 for up to 72 h
of 2 μg·kg−1·min−1 for up to 18–24 h. A second • PCI: 180-μg/kg bolus followed by infusion of
180-μg/kg bolus given 10 min after the first bolus 1 μg·kg−1·min−1 for up to 18–24 h. A second 180-μg/kg
bolus given 10 min after the first bolus
Contraindicated in dialysis patients
Fondaparinux26,27 75% • STEMI patients receiving fibrinolytic therapy: CrCl < 30 mL/min:
2.5 mg IV followed by 2.5 mg SC daily starting • Avoid use
the following day
• UA/NSTEMI: 2.5 mg SC daily
(Continued )
Washam et al   Pharmacotherapy in CKD Patients With ACS   1127

Table 1.  Continued


Renal
Medication Elimination Dose in Patients Without CKD Dose Adjustment in CKD

Unfractionated NS • UA/NSTEMI (initial dosing): Bolus of 60 U/kg No adjustment recommended


heparin26,27 (maximum 4000 U) followed by an infusion of
12 U·kg−1·h−1 (maximum 1000 U/h) to maintain
aPTT at 1.5–2.0 times control
• STEMI patients receiving fibrinolytic therapy:
Bolus of 60 U/kg (maximum 4000 U/kg) followed
by an infusion of 12 U·kg−1·h−1 (maximum 1000 U)
initially, adjusted to maintain aPTT at 1.5–2.0 times
control for 48 h or until revascularization.
Tirofiban26 65% • PCI: 25 μg/kg IV over 3 min followed by an infusion CrCl ≤60 mL/min:
of 0.15 μg·kg−1·min−1 for up to 18 h post-PCI • P CI: 25 μg/kg IV over 3 min followed by an infusion
of 0.075 μg·kg−1·min−1 for up to 18 h post-PCI
ACS indicates acute coronary syndrome; aPTT, activated partial thromboplastin time; CKD, chronic kidney disease; CrCl, creatinine clearance; IV, intravenous; NS,
not significant; NSTEMI, non–ST-segment–elevation myocardial infarction; PCI, percutaneous coronary intervention; SC, subcutaneous; STEMI, ST-segment–elevation
myocardial infarction; and UA, unstable angina.

and colleagues conducted an observational analysis to assess significant increase in mortality among patients with renal
the prognostic significance of baseline SCr in a study of 352 dysfunction (P<0.001) but no difference in major bleeding
STEMI patients receiving fibrinolytic therapy.31 In this study, (P=0.363). In 5549 Canadian ACS patients without ESRD who
there was no association between baseline SCr and TIMI flow survived to hospital discharge and were followed up for a mean
grade after fibrinolytic administration; however, there was a of 5.6 years, moderate (eGFR 30–59 mL·min−1·1.73 m−2) and

Table 2.  Summary of Fibrinolytic Studies in STEMI Patients With CKD


Study Study Design Population End Points Results
Gibson et al30 Subgroup analysis 16 635 patients received fibrinolytic therapy Mortality (30 d) A stepwise increase in 30-d mortality was seen
Downloaded from http://ahajournals.org by on January 29, 2019

of pooled data in a clinical trial and were divided into 4 Angiographic outcomes in patients with normal, mildly, moderately, and
from 4 trials categories according to renal function: (TIMI flow grade, corrected severely impaired renal function with rates of
normal (CrCl ≥90 mL/min, n=6062), mildly TIMI frame count) 2.2%, 5.2%, 13.8%, and 30.7%, respectively
impaired (CrCl 60–89 mL/min, n=6795), (P<0.0001).
moderately impaired (CrCl 30–59 mL/min, In patients who had angiographic assessment,
n=3514), and severely impaired (CrCl the rates of TIMI flow grade 2 or 3 at 90 min
<30 mL/min, n=264) were 80.7%, 80.2%, 85.5%, and 93.3%
(P=0.11 for trend) in the normal, mildly,
moderately, and severely impaired groups,
respectively.
ICH ICH rates in the 4 renal function categories were
0.6%, 0.8%, 1.8%, and 3.0%, respectively
(P<0.001 for trend)
Hobbach Observational 352 STEMI patients received fibrinolytic Mortality (30 d, 6 mo) 30-d mortality rates in patients in the normal
et al31 cohort therapy. Patients were divided into groups Reinfarction (30 d, 6 mo) and renal dysfunction groups were 3.4% and
based on SCr: normal (SCr <1.2 mg/dL, 16.1% (P<0.001), respectively. 30-d rates of
n=256) and renal dysfunction (SCr 1.3–2.8 reinfarction in the normal and renal dysfunction
mg/dL, n=87) groups were 3.4% and 3.6% (P=0.981),
respectively.
Major and minor bleeding Rates of major bleeding in the normal and
renal dysfunction groups were 2.6% and 4.6%
(P=0.363), respectively, whereas rates of minor
bleeding were 6.4% and 10.3% (P=0.224),
respectively.
Keough-Ryan Observational 5549 consecutive patients with ACS Mortality (mean follow-up Moderate and severe CKD were found to be
et al32 cohort (ICONS surviving to hospital discharge. Renal 5.6 y) independent predictors of mortality. Patients
registry) function classified into normal (>80; with severe CKD were less likely to receive
n=1430), mild CRI (60–80; n=2018), fibrinolytic therapy (OR, 0.55; 95% CI, 0.30–
moderate CRI (30–59; n=1795), and severe 0.98). The adjusted HR (95% CI) for mortality
CRI (<30; n=306) mL·min−1·1.73 m−2.* ESRD was 0.885 (0.81–0.97) for the use of fibrinolytic
patients were excluded from this analysis. therapy during hospitalization and 0.846
(0.79–0.91) for cardiac catheterization.
(Continued )
1128  Circulation  March 24, 2015

Table 2.  Continued


Study Study Design Population End Points Results

Medi et al33 Observational 12 532 patients with ST-segment Mortality (in-hospital and In this analysis, mortality increased as GFR
cohort (GRACE elevation or LBBB. Patients stratified 6 mo) decreased (P<0.001). The adjusted ORs
registry) into groups based on estimated GFR*: Reinfarction (95% CIs) for in-hospital mortality in patients
normal, ≥60 mL·min−1·1.73 m−2 (n=9082); receiving fibrinolysis vs patients receiving no
moderate, 30–59 mL·min−1·1.73 m−2 reperfusion were as follows: normal, 1.06
(2982); and severe renal dysfunction, (0.78–1.44); moderate, 1.35 (1.01–1.80);
<30 mL·min−1·1.73 m−2. severe renal dysfunction, 1.11 (0.57-2.14).
Major bleeding Compared with patients receiving no
reperfusion, the rates of major bleed with
fibrinolytic therapy in the moderate and
severe renal dysfunction groups were 2.5% vs
3.4% (P=0.11) and 4.2% vs 8.2% (P=0.12),
respectively
Dragu et al34 Observational 132 STEMI patients with renal failure Mortality (7 d, 30 d, 1 y) The unadjusted mortality rates at 7 d in the
cohort (ACSIS) (defined by a history or SCr >1.5 mg/dL 3 groups were as follows: fibrinolytic therapy,
on admission). 24 (18.2%) received 8% (2/24 patients); PCI, 26% (9/35 patients); and
fibrinolytic therapy, 35 (26.5%) received no reperfusion, 15% (11/73 patients; P=0.18).
primary PCI, and 73 (55.3%) received Unadjusted mortality rates were lower in the
no reperfusion. fibrinolytic therapy group at 30 d: 8%, 40%, and
30% (P=0.03), respectively. Bleeding rates were
not reported in this analysis
ACS indicates acute coronary syndrome; ACSIS, Acute Coronary Syndromes Israeli Survey; CI, confidence interval; CKD, chronic kidney disease; CrCl, creatinine
clearance; CRI, chronic renal insufficiency; ESRD, end-stage renal disease; GFR, glomerular filtration rate; GRACE, Global Registry of Acute Coronary Events; HR, hazard
ratio; ICH, intracranial hemorrhage; ICONS, Improved Cardiac Outcomes in Nova Scotia; LBBB, left bundle branch block; OR, odds ratio; PCI, percutaneous coronary
intervention; SCr, serum creatinine; STEMI, ST-segment–elevation myocardial infarction; and TIMI, Thrombolysis in Myocardial Infarction.
*GFR calculated by the modified Modification of Diet in Renal Disease equation.

severe (eGFR <30 mL·min−1·1.73 m−2) CKD were independent all patients received a fibrinolytic agent show increasing rates
Downloaded from http://ahajournals.org by on January 29, 2019

predictors of mortality.32 Factors associated with lower mortal- of intracranial hemorrhage with worsening renal function.30
ity included thrombolysis (hazard ratio [HR], 0.89; 95% con- This observation is noteworthy because current models for
fidence interval [CI], 0.81–0.97) and cardiac catheterization estimating intracranial hemorrhage risk with fibrinolytic ther-
(HR, 0.85; 95% CI, 0.79–0.91). Among 12 532 patients with apy do not include CKD as a risk factor.35,36 In spite of the lim-
ST-segment elevation or left bundle branch block enrolled in itations, taken collectively, the published data would support
the Global Registry of Acute Coronary Events (GRACE), in- that fibrinolytic therapy be considered as a treatment strategy
hospital mortality increased with worsening renal function for CKD patients presenting with STEMI when primary PCI
(P<0.001), and the use of reperfusion decreased with worsen- is not available. However, given the increased rates of intra-
ing renal function (P<0.001).33 Compared with no reperfusion, cranial hemorrhage observed with worsening renal function,
fibrinolytic therapy was not associated with in-hospital mortal- careful consideration of the benefits and risk of fibrinolytic
ity for patients with normal or severe renal dysfunction, but it therapy in this population is required.
was associated with increased mortality among patients with
moderate renal dysfunction (adjusted odds ratio [OR], 1.35;
Antiplatelet Therapy
95% CI, 1.01–1.80). A final observational analysis compared
reperfusion strategies among 132 STEMI patients with renal Aspirin
failure (defined by history or an SCr ≥1.5 mg/dL on admis- Current guidelines recommend aspirin should be initiated as
sion) enrolled in the Acute Coronary Syndromes Israeli Survey soon as an ACS is suspected and should be continued indefi-
(ACSIS).34 In this cohort, 24 patients (18.2%) received fibri- nitely, unless a contraindication develops.26,27 Given that
nolytic therapy, 35 (26.5%) were treated by primary PCI, and patients with renal insufficiency have an increased bleeding
73 (55.3%) received no reperfusion therapy. There was no sig- risk, there is some trepidation regarding the use of antiplatelet
nificant difference in mortality among the fibrinolytic, primary therapy in these patients. Although patients with CKD were
PCI, and no reperfusion groups at 7 days; however, at 30 days, excluded from most randomized trials of aspirin therapy in
mortality was lower among patients who received the fibrino- ACS, data on observational studies evaluating aspirin therapy
lytic strategy (8%) than among those with primary PCI (40%) in patients with renal impairment are shown in Table 3.
or no reperfusion (30%; P=0.03). The Antithrombotic Trialists’ Collaboration performed
In summary, although the above data suggest an increase in a meta-analysis of 287 randomized trials that included
adverse outcomes with worsening renal function, assessment 135 000 patients and compared various antiplatelet thera-
of the treatment effect of fibrinolytic therapy in the subgroup pies versus control.37 Some of those trials included patients
of patients with CKD is limited and variable. Data from a undergoing hemodialysis. Among those undergoing hemo-
pooled analysis of early trials of fibrinolytic therapy in which dialysis, antiplatelet therapy reduced the risk of serious
Washam et al   Pharmacotherapy in CKD Patients With ACS   1129

Table 3.  Summary of Aspirin Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
Antithrombotic Meta-analysis 287 studies involving 135 000 patients in Serious vascular event Absolute reduction in the risk of serious vascular
Trialists’ of randomized comparisons of antiplatelet therapy versus (nonfatal MI, nonfatal event in patients with previous MI was 36 (SE 5)
Collaboration37 trials control and 77 000 in comparisons of stroke, or vascular death per 1000 treated for 2 y and 38 (SE 5) per 1000
different antiplatelet regimens. The number treated for 1 mo with AMI. Among individuals
of hemodialysis patients who received undergoing hemodialysis, antiplatelet therapy
antiplatelet treatment was 1333, whereas produced a 41% (SE 16%) proportional reduction
the number of hemodialysis patients who in serious vascular events and no significant
received control was 1371. increase in extracranial bleeds (2% in antiplatelet
vs 2.3% in control).
Keough-Ryan Observational 5549 consecutive patients with ACS Mortality (mean follow-up A significant interaction was found between
et al32 cohort surviving to hospital discharge. Renal 5.6 y) kidney function and discharge aspirin therapy
function classified into normal, >80 with regard to mortality. The protective effect of
mL·min−1·1.73 m−2) n=1430; mild CRI, aspirin was attenuated with increasing degrees
60–80 mL·min−1·1.73 m−2 (n=2018); of renal dysfunction. The adjusted HR (95% CI)
moderate CRI, 30–59 mL·min−1·1.73 m−2 for aspirin overall was 0.904 (0.843–0.969);
(n=1795); and severe CRI, <30 (n=306) in mild CRI, it was 0.851 (0.921–1.128); in
mL·min−1·1.73 m−2 * moderate CRI, 1.029 (0.988–1.081); and in
severe CRI, 1.232 (1.024–1.117).
McCullough Observational, 1724 patients with STEMI. Renal function In-hospital mortality Adjusted RR reduction for the combination
et al21 prospective classified by quartile corrected creatinine of in-hospital aspirin and β-blocker was 80%,
cohort study clearance >81.5 mL/min (n=524), 63.1 74.9%, 69%, 64.3%, and 77.9% across the
to ≤81.5 (n=421), 46.2 to ≤63.1 (n=421), quartiles of corrected creatinine clearance,
≤46.2 not undergoing dialysis (n=310), respectively
and chronic dialysis (n=47)
Berger et al19 Observational, AMI patients: 145 740 patients without Mortality (30 d) Aspirin therapy was associated with a 50%
retrospective ESRD and 1025 patients with ESRD relative reduction in mortality in those receiving
analysis of CCP receiving dialysis dialysis (P<0.001) and a 63% relative reduction
and USRDS among those without ESRD (P<0.001)
Yan et al38 Observational, 3510 patients hospitalized for ACS with 1-y Survival Aspirin was associated with improved 1-y
Downloaded from http://ahajournals.org by on January 29, 2019

prospective normal renal function (CrCl† ≥90 mL/min; survival to a similar extent among those with
cohort study n=1152), mild renal dysfunction (CrCl normal and impaired renal function. Discharge
60–89 mL/min; n=1253), moderate aspirin use adjusted OR 0.43 (95% CI, 0.31–
renal dysfunction (CrCl 30–59 mL/min; 0.60; P<0.001; P=0.15 for heterogeneity across
n=944), and severe renal dysfunction CrCl <60 vs ≥60 mL/min)
(CrCl <30 mL/min; n=161)
Wright et al2 Observational, 3106 patients with AMI with no renal Short- and long-term Aspirin within 24 h of admission was
retrospective disease (n=1320), mild CRI (CrCl >50 survival stratified by CrCl associated with a significant reduction in
cohort study mL/min but ≤75 mL/min; n=860), in-hospital death adjusted OR 0.4 (95% CI,
moderate renal dysfunction (CrCl >35 0.3–0.6; P<0.001), and discharge aspirin
mL/min but ≤50 mL/min; n=491), severe was associated with improved postdischarge
renal insufficiency (CrCl† <35 mL/min; survival across the spectrum of renal failure
n=391), or ESRD (n=44) (adjusted HR, 0.7; 95% CI, 0.5–0.8; P<0.001)
Sciahbasi et al39 Retrospective 595 patients with AMI, 404 with normal Ratio of NSTEMI to STEMI Prior therapy with aspirin or statins was
review renal function, and 191 with impaired renal patients associated with increased ratio of NSTEMI to
function (GFR* ≤60 mL·min−1·1.73 m−2) STEMI in the overall population and in those
with impaired renal function. Adjusted OR (95%
CI) for STEMI with prior aspirin or statin therapy
in the overall population was 0.8 (0.65–0.93;
P=0.008) and in those with renal impairment,
0.5 (0.2–1.0; P=0.05)
ACS, acute coronary syndrome; AMI, acute myocardial infarction; CCP, Cooperative Cardiovascular Project; CI, confidence interval; CKD, chronic kidney disease; CrCl,
creatinine clearance; CRI, chronic renal insufficiency; ESRD, end-stage renal disease; GFR, glomerular filtration rate; HR, hazard ratio; MI, myocardial infarction; NSTEMI,
non–ST-segment–elevation myocardial infarction; OR, odds ratio; RR, relative risk; SE, standard error; STEMI, ST-segment–elevation myocardial infarction; and USRDS,
United States Renal Data System.
*GFR calculated by the modified Modification of Diet in Renal Disease equation.
†CrCl calculated via Cockcroft-Gault equation.

vascular events (nonfatal MI, nonfatal stroke, or vascular Most of the published observational data show similar ben-
death) by 41% (standard error, 16%). There was no signifi- efits of aspirin therapy in ACS patients across the spectrum of
cant increase in extracranial bleeds, although the absolute renal function (Table 3). However, one study did find a signifi-
number was small (2% in antiplatelet-treated patients versus cant interaction between discharge aspirin therapy and renal
2.3% in control subjects).37 function, with an attenuated benefit with increasing degree of
1130  Circulation  March 24, 2015

renal dysfunction.32 Although not conducted in ACS patients Optimizing Platelet Inhibition With Prasugrel–Thrombolysis
per se, the United Kingdom Heart and Renal Protection Study in Myocardial Infarction (TRITON–TIMI 38) randomized
and the Dialysis Outcomes and Practice Patterns Study both subjects who presented with moderate- to high-risk ACS with
showed no increased bleeding risk with aspirin therapy among scheduled PCI to either prasugrel or clopidogrel. Within this
patients receiving hemodialysis, which provides further support study, the risk reduction associated with prasugrel versus
for the safety of aspirin in patients with CKD.40,41 Collectively, clopidogrel was 20% among those with CrCl ≥60 mL/min
the available data suggest that aspirin therapy is safe and effec- and 14% among those with CrCl <60 mL/min.49 The Study of
tive in ACS patients with CKD and should be used in these Platelet Inhibition and Patient Outcomes (PLATO) random-
patients to reduce the risk of death and vascular events. ized patients admitted to the hospital with an ACS to treatment
with ticagrelor or clopidogrel. The HRs (95% CIs) for ticagre-
Clopidogrel, Prasugrel, and Ticagrelor lor versus clopidogrel for the primary composite outcome
Current guidelines recommend the use of a P2Y12 receptor were 0.90 (0.79–1.02) among subjects with CrCl ≥60 mL/min
inhibitor across the spectrum of ACS presentations.26,42 Data and 0.77 (0.65–0.90) among those with CrCl <60 mL/min.50,51
evaluating P2Y12 receptor inhibitors in patients with ESRD The HRs (95% CIs) for ticagrelor versus clopidogrel for major
are limited, and such information is available predominantly bleeding events were 1.08 (0.96–1.22) among patients with
for individuals with moderate or no CKD. Data on the use of CrCl >60 mL/min and 1.07 (0.88–1.30) among those with
P2Y12 receptor inhibitors in ACS patients with CKD are sum- CrCl <60 mL/min. Thus, the efficacy associated with prasu-
marized in Table 4. The Clopidogrel in Unstable Angina to grel and ticagrelor compared with clopidogrel was apparent
Prevent Recurrent Events (CURE) trial randomized patients among subjects with reduced and normal renal function.
with an ACS without ST-segment elevation to either a 300- In summary, randomized placebo-controlled trial data
mg loading dose of clopidogrel followed by 75 mg per day on the use of clopidogrel in ACS patients with CKD have
or placebo. Based on tertiles of renal function, the rela- been derived primarily from patients not undergoing an early
tive risks (RR) and 95% CIs for the primary composite out- invasive strategy or primary PCI.43,47 The lack of a treatment-­
come associated with clopidogrel versus placebo were 0.74 by–renal function interaction suggests clopidogrel should
(0.60–0.93) in the upper third, 0.68 (0.56–0.84) in the middle be considered as a treatment option in this population. In
third, and 0.89 (0.76–1.05) in the lower third, with a Pinteraction addition, although the observed rates of bleeding have been
of 0.11.43,44 In the Clopidogrel for the Reduction of Events higher with clopidogrel than with placebo in CKD patients,
During Observation (CREDO, a trial of patients undergoing the lack of a treatment interaction suggests no significant
planned PCI or coronary angiogram)45,46 and the Clopidogrel increase in risk with the use of clopidogrel in ACS patients
Downloaded from http://ahajournals.org by on January 29, 2019

as Adjunctive Reperfusion Therapy–Thrombolysis in with CKD. The efficacy associated with prasugrel compared
Myocardial Infarction 28 (CLARITY-TIMI 28) trial (a trial with clopidogrel and the efficacy and safety associated with
of patients with STEMI),47,48 there was a qualitative decline ticagrelor compared with clopidogrel were evident in patients
in the efficacy of clopidogrel versus placebo as renal func- with and without CKD, and the data suggest these agents
tion worsened; the HRs (95% CIs) across renal function were should be considered in CKD patients who are not consid-
0.42 (0.26–0.69), 0.80 (0.51–1.25), and 1.42 (0.81–2.45) in ered to be at high risk of bleeding. However, patients with
CREDO and 0.6 (0.40–0.90), 0.6 (0.40–0.70), and 1.0 (0.70– ESRD have been excluded from the landmark trials of these
1.6) in CLARITY-TIMI 28. In a substudy of the Clopidogrel newer agents.49,50
for High Atherothrombotic Risk and Ischemic Stabilization,
Management, and Avoidance (CHARISMA) trial, among Glycoprotein IIb/IIIa Receptor Antagonists
patients with and without diabetic nephropathy, the HRs (95% The glycoprotein (GP) IIb/IIIa receptor antagonists have
CIs) for clopidogrel versus placebo were 0.9 (0.80–1.0) for been studied extensively in patients undergoing PCI and
those without diabetic nephropathy and 1.1 (0.80–1.6) for in patients presenting with ACS. In the setting of STEMI,
those with diabetic nephropathy.52,53 recent guidelines give a Class IIa recommendation for the
In terms of safety, more bleeding occurred with clopido- use of the GP IIb/IIIa receptor antagonists at the time of
grel than placebo overall; however, there was no signifi- primary PCI in patients receiving unfractionated heparin
cant interaction based on renal function in CURE, CREDO, (UFH).26 Among patients presenting with unstable angina
or CLARITY-TIMI 28. Within the CHARISMA analysis, (UA)/NSTEMI with medium- or high-risk features in whom
the frequency of severe bleeding according to the Global an initial invasive strategy is selected, current recommenda-
Utilization of Streptokinase and tPA for Occluded Arteries tions for the use of GP IIb/IIIa receptor antagonists include
(GUSTO) definition among patients with diabetic nephropa- the option for upstream initiation or initiation at the time
thy was nonsignificantly higher with clopidogrel than with of PCI.42 Additionally, the guidelines favor a selective strat-
placebo (2.6% versus 1.5%, P=0.075). In patients without dia- egy of upstream use of GP IIb/IIIa receptor antagonists, and
betic nephropathy, there was no difference in GUSTO severe the use of these agents as part of an upstream triple-anti-
bleeding between patients randomized to clopidogrel versus platelet therapy regimen may not be supported when there
placebo (1.5% versus 1.3%, P=0.28).53 is a concern for increased bleeding risk.42 Of the 3 agents
Prasugrel and ticagrelor are P2Y12 inhibitors that exhibit currently available in the United States, eptifibatide and
a more rapid onset, higher degrees of platelet inhibi- tirofiban are dependent on renal clearance for elimination,
tion, and less interpatient variability than clopidogrel. The and dose adjustment is required for the 2 agents in patients
Trial to Assess Improvement in Therapeutic Outcomes by with CrCl <50 mL/min and CrCl ≤60 mL/min (Table 1),
Washam et al   Pharmacotherapy in CKD Patients With ACS   1131

Table 4.   Summary of Clopidogrel, Prasugrel, and Ticagrelor Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
CURE, Subgroup analysis 12 253 patients who presented with ACS Cardiovascular death, MI, Based on tertiles of renal function,† the RRs
200143,44 of an RCT without ST-segment elevation randomized or stroke through 1 y (95% CIs) for clopidogrel vs placebo were 0.74
to clopidogrel or placebo in addition to (0.60–0.93) for the upper third, 0.68 (0.56–
aspirin. Patients grouped based on tertiles 0.84) for the middle third, and 0.89 (0.76–1.05)
of eGFR*: upper tertile (>81.3 mL/min), for the lower third (Pinteraction=0.11).
middle tertile (64–81.2 mL/min), lower Major or life-threatening The RRs (95% CIs) for major or life-threatening
tertile (<64 mL/min). An eGFR <30 mL/min bleeding bleeding for clopidogrel vs placebo were 1.83
was noted in 1.8% (n=224) of patients. (1.23–2.73) for the upper third, 1.4 (0.97–2.02)
for the middle third, and 1.12 (0.83–1.51) for
the lower third.
CREDO, Subgroup analysis 2002 patients referred for a planned PCI Death, MI, or stroke Based on estimated CrCl† (>90 [normal],
200345,46 of an RCT or coronary angiogram randomized to through 1 y 60–89 [mild CKD], <60 mL/min [moderate
clopidogrel initiated with a 300 mg load CKD]), the HRs (95% CIs) for clopidogrel vs
followed by 75 mg daily vs clopidogrel 75 placebo were 0.42 (0.26–0.69) for the normal
mg daily through 28 d. Patients grouped group, 0.80 (0.51–1.25) for mild CKD, and 1.41
according to CrCl: >90 (n=999), 60–89 (0.81–2.45) for moderate CKD.
(n=672), <60 mL/min (n=331) Major bleeding through 1 y The RRs (95% CIs) for clopidogrel vs placebo
were 1.17 (0.74–1.84) for the normal group,
1.59 (0.97–2.62) for mild CKD, and 1.12 (0.51–
2.48) for moderate CKD.
CLARITY-TIMI Subgroup analysis 3252 patients who presented within 12 h Death, MI, or TIMI 0/1 The HRs (95% CIs) for clopidogrel vs placebo
28, 200547,48 of an RCT after the onset of an ST-segment elevation flow through angiography were 0.6 (0.4–0.9) for the normal group, 0.6
MI who received fibrinolytic therapy or day 8 (0.4–0.7) for mild CKD, and 1.0 (0.7–1.6) for
randomized to clopidogrel vs placebo. moderate CKD (P interaction=0.09).
Patients were stratified based on normal TIMI major or minor bleeding The adjusted ORs (95% CIs) for clopidogrel
(≥90) (n=841), mild (60– 89)(n=1,897) and at 30 d vs placebo were 1.7 (0.5-5.3) for the normal
moderate (<60 mL/min/1.73 m2) (n=514) group, 1.3 (0.8–2.2) for the mild group, and
reductions in GFR.* 1.6 (0.7–3.7) for the group with moderate CKD
(Pinteraction=0.94).
Downloaded from http://ahajournals.org by on January 29, 2019

TRITON-TIMI Subgroup analysis 13 608 patients with moderate- to high-risk Cardiovascular death, MI, The reduction in risk with prasugrel vs
38, 200749 of an RCT ACS with scheduled PCI randomized to or stroke through 15 mo clopidogrel was 20% among subjects with
prasugrel vs clopidogrel. Patients grouped CrCl† ≥60 mL/min and 14% among those with
according to CrCl†: >60 mL/min (11 890) CrCl <60 mL/min.
and <60 mL/min (n=1490)
PLATO, Subgroup analysis 15 202 patients admitted to the hospital Cardiovascular death, MI, The HRs (95% CIs) for ticagrelor vs clopidogrel
200950,51 of an RCT with an ACS randomized to ticagrelor or stroke through 12 mo were 0.90 (0.79–1.02) among subjects with
vs clopidogrel. Patients were grouped CrCl† ≥60 mL/min and 0.77 (0.65–0.90) among
according to CrCl†: ≥60 mL/min those with CrCl <60 mL/min (Pinteraction=0.13).
(n=11 965) and <60 mL/min (n=3237) The HR (95% CI) in those with CrCl <30 mL/min
(n=214) was 0.77 (0.49–1.30).
Major bleeding In patients with CrCl <60 mL/min, the HR
(95% CI) for major bleeding events for
ticagrelor compared with clopidogrel was
1.07 (0.88–1.30).
ACS indicates acute coronary syndrome; CI, confidence interval; CKD, chronic kidney disease; CLARITY-TIMI 28, Clopidogrel as Adjunctive Reperfusion Therapy–
Thrombolysis in Myocardial Infarction 28; CrCl, creatinine clearance; CREDO, Clopidogrel for the Reduction of Events During Observation; CURE, Clopidogrel in Unstable
Angina to Prevent Recurrent Events; eGFR, estimated glomerular filtration rate; HR, hazard ratio; MI, myocardial infarction; OR, odds ratio; PCI, percutaneous coronary
intervention; PLATO, Study of Platelet Inhibition and Patient Outcomes; RCT, randomized controlled trial; RR, relative risk; TIMI, Thrombolysis in Myocardial Infarction;
and TRITON-TIMI 38, Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition With Prasugrel-Thrombolysis in Myocardial Infarction 38.
*Calculated by modified Modification of Diet in Renal Disease equation.
†Calculated via Cockcroft-Gault equation.

respectively.54,55 In addition, eptifibatide is contraindicated in When used at the time of PCI in ACS patients with CKD,
patients requiring dialysis.54 outcomes with the use of GP IIb/IIIa receptor antagonists
Abciximab is cleared via the reticuloendothelial system, have been variable. A subgroup analysis of the Enhanced
and no current recommendations exist for dose adjustment for Suppression of the Platelet IIb/IIIa Receptor With Integrilin
patients with CKD. Data from randomized trials and observa- Therapy (ESPRIT) trial showed the treatment effect of
tional studies on the clinical outcomes of ACS patients with eptifibatide was maintained among those with CrCl <60
CKD receiving a GP IIb/IIIa receptor antagonist are summa- mL/min, and the presence of CKD was not associated with
rized in Table 5. an increased risk of bleeding with eptifibatide therapy.58
1132  Circulation  March 24, 2015

Observational cohorts of ACS patients receiving GP IIb/IIIa frequency of death or MI (HR, 1.03; 95% CI, 0.97–1.08).
receptor antagonists at the time of PCI have provided addi- A second observational study categorized patients undergo-
tional insight into use in patients with CKD. Best et al57 ing PCI into 5 strata by CrCl (≥90 mL/min, 60–89 mL/min,
assessed patients undergoing PCI in a large registry that 30–59 mL/min, <30 mL/min, and requiring dialysis).59 After
divided patients into 3 categories according to creatinine controlling for CrCl stratum, GP IIb/IIIa receptor antagonist
clearance: >70, 50–69, or <50 mL/min. Among patients use was associated with a lower risk of in-hospital mortal-
receiving abciximab, the interaction between CrCl and major ity (OR, 0.34; 95% CI, 0.12–0.98) and an increased risk of
bleeding was not statistically significant. Additionally, no a major bleeding event (OR, 2.13; 95% CI, 1.39–3.27). A
interaction was seen between CrCl and abciximab for the final observational study reported the clinical outcomes of

Table 5.  Summary of GP IIb/IIIa Receptor Antagonist Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
PRISM-PLUS56 Post hoc analysis 1537 UA/NSTEMI patients randomized to 7-d death/MI/refractory Decreasing renal function was strongly
of an RCT receive tirofiban or placebo. Renal function ischemia (primary associated with adverse outcomes,
classified by CrCl*: >75 mL/min (n=572), composite) however, there was no evidence of
60–75 mL/min (n=354), 30–60 mL/min treatment-by-CrCl interaction. The
(n=571), <30 mL/min (n=40). Patients incidence of the composite end point in the
with SCr ≥2.5 mg/dL were excluded from tirofiban and placebo groups at 7 d was
the PRISM-PLUS trial. 35% vs 45% in patients with CrCl <30
mL/min, 17.9% vs 23.8% in patients with
CrCl 30–60 mL/min, 13.9% vs 15.5% in
patients with CrCl 60–75 mL/min, and 6.8%
vs 12.3% in patients with CrCl >75 mL/min.
TIMI major bleeding, No incremental risk for bleeding was
minor bleeding observed with tirofiban therapy among
the lowest CrCl categories. No TIMI
major bleeding events occurred in either
treatment group in the CrCl <30 mL/min
category; for the 30–60 mL/min category,
the number of major bleeding events was
Downloaded from http://ahajournals.org by on January 29, 2019

4 (1.4%) in the placebo group vs 5 (1.8%)


in the tirofiban group.
Best et al57 Observational cohort 4158 patients undergoing PCI (indication Composite of death or MI No interaction was seen between CrCl and
for PCI was UA in 71% of patients in each abciximab for the frequency of death or MI
group and MI in the previous 7 d in 26% of (HR, 1.03; 95% CI, 0.97–1.08).
patients in the abciximab group and 15% of
patients in the no abciximab group). Renal
function classified based on CrCl*: >70
mL/min (n=647 who received abciximab
and n=1472 who did not), 50–69 mL/min
(n=367 who received abciximab and 820
who did not), and <50 mL/min (n=229 who
received abciximab and 585 who did not).
Major bleeding, minor CKD was associated with increased
bleeding (follow-up 10 d) bleeding complications. In patients who
received abciximab, the interaction between
CrCl and major bleeding (OR, 1.18; 95%
CI, 0.99–1.39) as well as CrCl and minor
bleeding (OR, 1.01; 95% CI, 0.83–1.23) did
not reach statistical significance.
ESPRIT58 Subgroup analysis Patients randomized to eptifibatide or Composite of death, MI, The adjusted ORs (95% CIs) for the effect
of an RCT placebo at the time of PCI. 2044 of 2064 TVR, or thrombotic bailout of eptifibatide on the primary outcome
patients had creatinine data available. assessed at 48 h remained for those with lower CrCl
A total of 1755 patients had CrCl* (60 mL/min) 0.52 (0.33–0.81) compared
≥60 mL/min, and 289 patients had CrCl with those with higher CrCl (90 mL/min)
<60 mL/min. Patients with SCr >4 mg/dL 0.64 (0.46–0.89).
were excluded. Indication for PCI was UA
in 44% of patients with CrCl ≥60 mL/min
and 54% in those with CrCl <60 mL/min.
Major bleeding, minor Lower CrCl was not associated with an
bleeding increased risk of bleeding with eptifibatide
therapy (Pinteraction=0.791).
(Continued )
Washam et al   Pharmacotherapy in CKD Patients With ACS   1133

Table 5.  Continued


Study Study Design Population End Points Results

Freeman et al59 Observational, 889 patients with ACS, including 310 In-hospital mortality Although worsening CrCl stratum was a
cohort study patients with renal insufficiency defined predictor of in-hospital mortality, a lower
as CrCl* <60 mL/min (222 patients had risk of in-hospital mortality was seen
CrCl of 30–59 mL/min, 63 had CrCl of with the use of GP IIb/IIIa antagonist after
<30 mL/min, and 25 patients were controlling for CrCl (OR, 0.34; 95% CI,
dialysis dependent). 312 of the 889 0.12-0.98).
patients enrolled received a GP IIb/IIIa Major bleeding events The use of GP IIb/IIIa antagonist was
antagonist. associated with an increase in major
bleeding events (OR, 2.13; 95% CI,
1.39–3.27).
Frilling et al60 Single-center registry 1040 patients including 44 with renal MACCE: death, MI, stroke, No statistically significant differences
insufficiency (defined as SCr ≥1.3 mg/dL) emergency aortocoronary were seen in MACCE rates between
undergoing PCI who received abciximab. bypass operation or PCI groups. In-hospital mortality occurred in
The indication for PCI was ACS in 718 of 4.5% of patients with renal insufficiency
996 patients without renal insufficiency vs 1.9% of patients without renal
and 35 of 44 patients with renal insufficiency (P=0.223). Nonfatal
insufficiency. MACCE was reported in 4.5% and 6.7%,
respectively (P=0.569).
Major and minor bleeding Major bleeding occurred in 4.5% of patients
events with renal insufficiency vs 0.6% of those
without renal insufficiency (P=0.003). No
differences were seen in the rates of minor
bleeding events.
TARGET61 Subgroup analysis 4623 patients undergoing PCI randomized Composite of all-cause Ischemic events occurred more frequently
of an RCT to abciximab or tirofiban were grouped mortality, MI, and TVR in patients with lower CrCls. There was no
into CrCl* quartiles (>114, 90–114, at 30 d interaction between the GP IIb/IIIa receptor
70–90, <70 mL/min). Patients with antagonist used, CrCl, and ischemic or
SCr >2.5 mg/dL were excluded from bleeding outcomes. In patients with CrCl
the TARGET trial. The indication for PCI <70 mL/min, the primary composite
Downloaded from http://ahajournals.org by on January 29, 2019

was ACS in 63% of patients. occurred in 6% of the abciximab group and


8.7% of the tirofiban group (P=0.74).
Major bleeding, minor Bleeding events occurred more frequently
bleeding in patients with lower CrCls. Significant
treatment differences in major bleeding
were not detected. In patients with CrCl
<70 mL/min, increased rates of bleeding
were observed with abciximab vs tirofiban
(7.2% vs 3.4%; P=0.004).
EARLY ACS62 Subgroup analysis 8708 patients with UA/NSTEMI were Primary composite: In patients with CrCl <50 mL/min: The
of an RCT randomized to early eptifibatide or a death, MI, recurrent primary outcome occurred in 12.5% of
strategy of delayed provision eptifibatide. ischemia requiring urgent those receiving early eptifibatide compared
A total of 1632 patients had a baseline revascularization, or to 11.7% receiving a delayed provisional
CrCl* <50 ­mL/min. Patients requiring thrombotic bailout at 96 h eptifibatide strategy (OR (95% CI) 1.08
dialysis within the past month were Death or MI at 30 d (0.80–1.45)). The rates of death or MI at
excluded. 30 d were 15.6% in the early eptifibatide
group vs 15.1% in the delayed provisional
eptifibatide group (OR (95% CI) 1.03
(0.79–1.35)).
Rates of non-CABG The rates of TIMI major bleeding (2.4% vs
related TIMI major 0.9%; OR, 2.91; 95% CI, 1.22–6.91) and
bleeding and GUSTO GUSTO severe/moderate bleeding (9.8%
severe/moderate bleeding vs 6.6%; OR, 1.52; 95% CI, 1.06-2.18)
were higher in the group receiving the early
eptifibatide strategy.
ACS indicates acute coronary syndrome; CABG, coronary artery bypass grafting; CI, confidence interval; CKD, chronic kidney disease; CrCl, creatinine clearance;
EARLY ACS, Early Glycoprotein IIb/IIIa Inhibition in Non-ST-Segment Elevation Acute Coronary Syndrome; ESPRIT, Enhanced Suppression of the Platelet IIb/IIIa Receptor
With Integrilin Therapy; GP, glycoprotein; GUSTO, Global Utilization of Streptokinase and tPA for Occluded Arteries; HR, hazard ratio; MACCE, major adverse cardiac or
cerebrovascular event; MI, myocardial infarction; NSTEMI, non–ST-segment–elevation myocardial infarction; OR, odds ratio; PCI, percutaneous coronary intervention;
PRISM-PLUS, Platelet Receptor Inhibition in Ischemic Syndrome Management in Patients Limited by Unstable Signs and Symptoms; RCT, randomized controlled trial;
SCr, serum creatinine; TARGET, Do Tirofiban and ReoPro Give Similar Efficacy Outcome; TIMI, Thrombolysis in Myocardial Infarction; TVR, target-vessel revascularization;
and UA, unstable angina.
*Calculated via Cockcroft-Gault equation.
1134  Circulation  March 24, 2015

patients undergoing PCI who received abciximab.60 Of the important that clinicians follow dosing recommendations for
1040 patients included, 44 were classified as having renal eptifibatide and tirofiban when either of these agents is used.
insufficiency. The authors reported no significant differences
in the rates of in-hospital mortality or nonfatal major adverse Anticoagulants
cardiac events among patients with renal insufficiency com-
pared with those without, although major bleeding occurred Unfractionated Heparin
more frequently among patients with renal insufficiency UFH has been a mainstay in the treatment of ACS for several
(4.5% versus 0.6%; P=0.003). Although limited data are decades.63 Current guidelines recommend UFH as an antico-
available on the comparative safety and effectiveness of dif- agulant option across the spectrum of ACS presentations.26,42
ferent GP IIb/IIIa receptor antagonists among patients with Once administered, the primary route of UFH elimination is
CKD, a subgroup analysis of the Do Tirofiban and ReoPro via the reticuloendothelial system, with renal clearance being
Give Similar Efficacy Outcome (TARGET) trial compared a minor route for elimination.64 Few data are available from
the outcomes of patients with renal insufficiency undergoing early placebo-controlled trials on the treatment effect of UFH
PCI who received either abciximab or tirofiban.61 The 4623 therapy in CKD patients presenting with ACS. In addition,
patients with a baseline SCr level available were divided into given that UFH has often been the standard anticoagulant
CrCl quartiles (<70, 70–90, 90–114, and >114 mL/min). with which newer agents have been compared, the outcome
Although the rates of both ischemic and bleeding events data for UFH use in CKD patients will be discussed in the
were higher among patients with lower creatinine clear- sections below.
ances, there was no interaction between the assigned GP
Low-Molecular-Weight Heparin
IIb/IIIa receptor antagonist, CrCl, and clinical outcome.
An analysis of the Platelet Receptor Inhibition in Ischemic Enoxaparin
Syndrome Management in Patients Limited by Unstable Signs Enoxaparin is the most widely studied low-molecular-weight
and Symptoms (PRISM-PLUS) trial provided outcome data heparin (LMWH) in the setting of ACS. Current guidelines
on the upstream use of GP IIb/IIIa receptor antagonists among recommend enoxaparin as an anticoagulant option in UA/
patients presenting with ACS.56 This analysis showed tirofi- NSTEMI patients being managed with either an early inva-
ban therapy to be effective in reducing ischemic events, with sive (Class I recommendation) or initial conservative (Class I
no evidence of treatment-by-CrCl interaction. Additionally, recommendation) strategy.42 For patients presenting with
although decreasing renal function (P<0.001) and tirofiban STEMI, current guidelines recommend enoxaparin as an
(P<0.001) were each associated with an increased risk for adjunctive anticoagulant option in conjunction with fibrino-
Downloaded from http://ahajournals.org by on January 29, 2019

bleeding events, tirofiban therapy was not associated with an lytic therapy (Class I recommendation).26 Enoxaparin elimi-
incremental increase in the risk for hemorrhage among those nation is largely dependent on renal function, with ≈40% of
with CKD. A subgroup analysis of the Early Glycoprotein IIb/ a dose being eliminated by glomerular filtration. The current
IIIa Inhibition in Non-ST-Segment Elevation Acute Coronary US Food and Drug Administration–approved dose for enoxa-
Syndrome (EARLY ACS) trial provided a comparative assess- parin in ACS patients with CrCl <30 mL/min is 1 mg/kg sub-
ment of the early versus delayed provisional use of eptifibatide cutaneously every 24 hours. However, patients with CrCl <30
among patients with CKD presenting with non–ST-segment mL/min have been routinely excluded from randomized trials
elevation ACS in whom coronary angiography was planned.62 of enoxaparin in ACS; therefore, limited data are available
This was the first large-scale randomized trial of eptifiba- from randomized controlled trials on the use of enoxaparin
tide that used the currently recommended dosing regimen in this population. Data from randomized trials and observa-
of 2 μg·kg−1·min−1 for patients with CrCl ≥50 mL/min and tional studies on the use of enoxaparin in ACS patients with
1 μg·kg−1·min−1 for patients with CrCl <50 mL/min, with CKD are shown in Table 6.
patients requiring dialysis excluded from the trial. Among A number of studies have shown increased anti-Xa activ-
patients with CrCl <50 mL/min, early eptifibatide compared ity in ACS patients with renal insufficiency receiving thera-
with a delayed provisional eptifibatide strategy was not asso- peutic doses of enoxaparin. In a substudy performed in 445
ciated with a reduction in the composite ischemic end point(s) ACS patients enrolled in the TIMI 11A trial, the effect of renal
at 96 hours or at 30 days. Among patients with CrCl <50 mL/ function and other patient characteristics on the pharmacoki-
min, rates of non–coronary artery bypass graft–related TIMI netics and pharmacodynamics of enoxaparin was examined.70
major bleeding and GUSTO severe/moderate bleeding were In this analysis, CrCl was the most influential factor on phar-
significantly higher with the early eptifibatide strategy. macokinetic and pharmacodynamic parameters of enoxa-
In summary, data on the use of GP IIb/IIIa receptor antago- parin. Patients with CrCl <40 mL/min had higher peak and
nists in CKD patients with ACS indicate a reduction in isch- trough anti-Xa activity than patients with CrCl ≥40 mL/min
emic events and a variable but overall increase in the risk of and were more likely to have a major hemorrhagic event.70
bleeding events. In addition, in a recent trial comparing a strat- Several clinical trials evaluating the use of enoxaparin in UA/
egy of early versus delayed provisional GP IIb/IIIa receptor NSTEMI patients have provided data on the outcomes of
antagonist therapy in UA/NSTEMI patients, no reduction in CKD patients receiving enoxaparin or UFH. A pooled anal-
ischemic events and an increase in bleeding events were seen ysis of CKD patients with severe renal impairment (defined
with the early strategy in patients with CKD.62 as CrCl ≤30 mL/min) enrolled in the Efficacy and Safety of
Although the existing data do not support a preferred GP Subcutaneous Enoxaparin in Non-Q-Wave Coronary Events
IIb/IIIa receptor antagonist for use in patients with CKD, it is (ESSENCE) and TIMI 11B trials was performed to assess
Washam et al   Pharmacotherapy in CKD Patients With ACS   1135

Table 6.  Summary of Enoxaparin Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
ESSENCE- Pooled 7081 NSTE ACS patients enrolled in ESSENCE All-cause death, In patients with CrCl <30 mL/min, the composite
TIMI 11B65 subgroup (n=3171) and TIMI 11B (n=3910) randomized recurrent MI, and primary outcome occurred in 32.4% of UFH
analysis of 2 to enoxaparin or UFH. CrCl ≤30 mL/min was urgent revas­ patients and 18.8% of enoxaparin patients (OR,
RCTs an exclusion in 1 trial (ESSENCE), whereas cularization at 43 d 0.52; 95% CI, 0.23–1.19).
SCr >2.0 mg/dL was an exclusion in the other Major hemorrhage Major hemorrhage occurred in 5.9% of UFH
(TIMI 11B). Severe renal impairment (CrCl* ≤30 patients and 7.5% of enoxaparin patients (OR,
mL/min) was found in 74 UFH-treated patients 1.53; 95% CI, 0.37–6.32).
and 69 patients receiving enoxaparin (2%). The
enoxaparin dose was 1 mg/kg every 12 h.
Collet et al66 Observational 11 881 patients presenting with UA/NSTEMI. Mortality (30 d) Worsening renal function was an independent
cohort (GRACE Patients were divided into 3 groups based on predictor of 30-d mortality and in-hospital
registry) CrCl*: >60 mL/min (n=7194), 31–60 mL/min major bleeding. Rates of 30-d mortality were
(n=3705), ≤30 mL/min (n=982). In the 3 renal significantly lower with LMWH alone than with
function groups, LMWH was used in 30%, 31%, UFH alone in patients with CrCl >60 mL/min and
and 30%, respectively, whereas UFH was used in those with CrCl 31–60 mL/min. No significant
in 22%, 24%, and 28%, respectively. difference was seen in patients with CrCl <30
mL/min (15.4% vs 18.6%, respectively).
In-hospital major Rates of in-hospital major bleeding were significantly
bleeding lower with LMWH alone than with UFH alone in
patients with normal and moderate renal dysfunction.
No significant difference was seen in patients with
CrCl <30 mL/min (5.9% vs 9.3%, respectively).
ExTRACT-TIMI Double blind 20 479 patients with STEMI receiving All-cause death or Adjusted OR (95% CI) for enoxaparin vs UFH
25 Trial67 RCT fibrinolytic therapy randomized to UFH or nonfatal recurrent MI comparison: Primary end point: 0.69 (0.56– 0.84)
enoxaparin. Patients with CrCl* <30 mL/min within 30 d for CrCl >90 mL/min, 0.78 (0.66–0.92) for CrCl
received a maintenance enoxaparin dose of >60 to 90 mL/min, 0.94 (0.78–1.12) for CrCl
1 mg/kg every 24 h. SCr >2.5 mg/dL for men 30–60 mL/min, and 0.74 (0.38– 1.44) for CrCl
and >2.0 mg/dL for women was an exclusion. <30 mL/min
Patients were divided into 4 groups based on Major bleeding Major bleeding: 1.49 (0.89–2.48) for CrCl >90
Downloaded from http://ahajournals.org by on January 29, 2019

CrCl*: >90 (n=7462), >60 to 90 (n=7203), mL/min, 1.91 (1.30– 2.82) for CrCl >60 to 90
30–60 (n=3671), and <30 mL/min (n=212). mL/min, 1.73 (1.11–2.70) for CrCl 30–60 mL/min,
and 3.60 (0.67–19.21) for CrCl <30 mL/min
SYNERGY68 Open-label 10 027 NSTE ACS patients randomized to All-cause death or No significant treatment-by-CrCl interaction term
RCT enoxaparin or UFH. Maintenance dose of nonfatal MI (30 d) was found for all treatment outcomes. 30-d death
enoxaparin was 1 mg/kg every 12 h. Early or MI in patients treated with UFH vs enoxaparin in
angiography intended (median time, 22 h). patients was 12.9% vs 12.7% for CrCl >60 mL/min,
Patients with CrCl <30 mL/min were to be 17.7% vs 17.0% for CrCl 30–59 mL/min, and 23.3%
excluded. Patients were grouped according to vs 25.7% for CrCl <30 mL/min
CrCl*: ≥60 mL/min (n=6950), 30–59 mL/min TIMI major bleeding TIMI major bleeding in patients treated with UFH
(n=2732), and <30 mL/min (n=156) vs enoxaparin in patients: CrCl >60 mL/min, 7.2%
vs 8.3%; CrCl 30–59 mL/min, 8.8% vs 11.2%;
CrCl <30 mL/min, 5.8% vs 10.0%
OASIS-569 Double-blind 20 078 patients randomized to fondaparinux Primary efficacy end HR (95% CI) for fondaparinux vs enoxaparin
RCT or enoxaparin. SCr level >3 mg/dL was an point: Death, MI, or comparison:
exclusion. Patients with CrCl* <30 mL/min refractory ischemia Efficacy end point at 30 d: 0.91 (0.74–1.12) for
received an enoxaparin maintenance dose of at 9 d eGFR ≥86 mL·min−1·1.73 m−2, 0.95 (0.76–1.18) for
1 mg/kg every 12 h. Patients were grouped and eGFR 71 to <86 mL·min−1·1.73 m−2, 1.10 (0.90–
analyzed in quartiles based on eGFR.† 1.33) for eGFR 58 to <71 mL·min−1·1.73 m−2, and
0.81 (0.69–0.96) for eGFR <58 mL/min/1.73 m2
Major bleeding Major bleeding at 30 d: 0.71 (0.53–0.96) for eGFR
≥86 mL·min−1·1.73 m−2, 0.87 (0.66–1.16) for
eGFR 71 to <86 mL·min−1·1.73 m−2, 0.74 (0.58–
0.95) for eGFR 58 to <71 mL·min−1·1.73 m−2, and
0.65 (0.52–0.80) for eGFR <58 mL·min−1·1.73 m−2
ACS indicates acute coronary syndrome; CI, confidence interval; CKD, chronic kidney disease; CrCl, creatinine clearance; eGFR, estimated glomerular filtration rate; ESSENCE,
Efficacy and Safety of Subcutaneous Enoxaparin in Non-Q-Wave Coronary Events; ExTRACT-TIMI 25, Enoxaparin and Thrombolysis Reperfusion for Acute Myocardial Infarction
Treatment–Thrombolysis In Myocardial Infarction 25; GRACE, Global Registry of Acute Coronary Events; HR, hazard ratio; LMWH, low-molecular-weight heparin; MI, myocardial
infarction; NSTE, non–ST-segment elevation; NSTEMI, non–ST-segment–elevation myocardial infarction; OASIS-5, Organization for the Assessment of Strategies for Ischemic
Syndromes 5; OR, odds ratio; RCT, randomized controlled trial; SCr, serum creatinine; STEMI, ST-segment–elevation myocardial infarction; SYNERGY, Superior Yield of the New
Strategy of Enoxaparin, Revascularization, and Glycoprotein IIb/IIIa Inhibitors; TIMI, Thrombolysis in Myocardial Infarction; UA, unstable angina; and UFH, unfractionated heparin.
*Cockcroft-Gault Formula.
†eGFR assessed by Modification of Diet in Renal Disease formula.
1136  Circulation  March 24, 2015

the treatment effects of enoxaparin and UFH.65 Severe renal that exclusively examined patients with renal dysfunction,
impairment was identified in ≈2% of patients in this pooled suggested that enoxaparin use was associated with a 2- to
analysis. There was no statistically significant difference 3-fold increase in major bleeding events when CrCl was
between enoxaparin and UFH with regard to the rates of <30 mL/min.71 In clinical practice, the use of enoxaparin in
occurrence of the primary composite end point (18.8% ver- ACS has been encumbered by challenges in correctly adjusting
sus 32.4%, respectively; P=0.12) or major hemorrhage (7.5% the dose of enoxaparin for creatinine clearance. In an analysis
versus 5.8%, respectively; P=0.56) among patients with CrCl of the CRUSADE Quality Improvement Initiative, enoxaparin
≤30 mL/min. The association of CKD with outcomes among was used in 40% of 33 094 patients with non–ST-segment–
patients enrolled in the Superior Yield of the New Strategy elevation ACS.72 Only 50% of patients treated with enoxapa-
of Enoxaparin, Revascularization, and Glycoprotein IIb/IIIa rin received the recommended dose according to their renal
Inhibitors (SYNERGY) trial who received either enoxapa- function, 18.7% received an excess dose, and 29.2% received
rin or UFH was also assessed.68 Patients were placed into a lower dose. An excess dose was associated with increased
3 groups based on CrCl: <30 mL/min (n=156), 30–59 mL/ risk of major bleeding and in-hospital mortality. Major bleed-
min (n=2732), and ≥60 mL/min (n=6950). No treatment- ing occurred in 14.2% and 7.3% of patients who received an
by-CrCl interaction was significant for the efficacy or safety excess and a recommended dose, respectively (adjusted OR,
outcomes, although the rates of TIMI major and GUSTO 1.43; 95% CI, 1.18–1.75). In-hospital death occurred in 5.6%
severe bleeding were numerically higher in patients with and 2.4% of patients who received an excess and a recom-
CrCl <30 mL/min and those with CrCl 30 to 59 mL/min. An mended dose, respectively (OR, 1.35; 95% CI, 1.03–1.77).
analysis of GRACE evaluated the association of CKD with
outcomes among patients with non–ST-segment–elevation
Factor Xa Inhibitors
ACS treated with either LMWH or UFH.66 Based on CrCl <30
mL/min (43 patients received UFH and 37 received LMWH), Fondaparinux
30 to 59 mL/min (70 patients received UFH and 49 received Fondaparinux is an indirect factor Xa inhibitor that has
LMWH), and >60 mL/min (50 patients received UFH and recently been evaluated in the management of patients pre-
45 received LMWH), results of this analysis showed that senting with ACS. Current guidelines for fondaparinux in ACS
LMWH was associated with lower 30-day mortality (4.2% patients include a Class I recommendation for fondaparinux
versus 6.2%; P<0.0001) and a lower rate of in-hospital major use as an adjunctive anticoagulant for STEMI patients receiv-
bleeds (2.1% versus 3.3%; P=0.0006), but the mortality and ing fibrinolytic therapy and a Class I recommendation for UA/
in-­hospital major bleeding benefit was not statistically signifi- NSTEMI patients being managed with either an invasive or
Downloaded from http://ahajournals.org by on January 29, 2019

cant in the group with CrCl <30 mL/min. conservative strategy.26,73 In addition, the guidelines recom-
The association of renal function with outcomes among mend fondaparinux as the preferred anticoagulant in UA/
fibrinolytic agent–treated STEMI patients receiving enoxapa- NSTEMI patients being managed with a conservative strategy
rin or UFH was evaluated in an analysis of the Enoxaparin and who have an increased risk of bleeding.73 Fondaparinux is pri-
Thrombolysis Reperfusion for Acute Myocardial Infarction marily excreted unchanged through renal elimination, and is
Treatment–Thrombolysis In Myocardial Infarction 25 contraindicated in the United States for patients with severe
(ExTRACT-TIMI 25) trial.67 In this trial, patients with CrCl CKD (CrCl <30 mL/min).74
<30 mL/min received a dose of enoxaparin of 1 mg/kg once The Organization for the Assessment of Strategies for
per day. On the basis of estimated CrCl <30 mL/min, 30 to Ischemic Syndromes (OASIS) 5 trial compared fondaparinux
60 mL/min, >60 to 90 mL/min, and >90 mL/min, there was to enoxaparin in 20  078 patients with non–ST-segment–­
a statistically significant benefit of enoxaparin on the primary elevation ACS randomized to fondaparinux 2.5 mg subcutane-
composite end point of death or nonfatal MI among patients ously once daily or enoxaparin 1 mg/kg subcutaneously twice
with CrCl >60 mL/min that was not seen in patients with daily (dose adjusted to 1 mg/kg once daily in patients with
CrCl 30 to 60 mL/min or in those with CrCl <30 mL/min. In CrCl <30 mL/min).75 Although patients with SCr >3 mg/dL
addition, there was an increase in the risk of major and minor were excluded, a specific analysis was designed to examine
bleeding events with enoxaparin treatment among patients efficacy and safety outcomes by quartiles of GFR (as esti-
with renal dysfunction (defined as CrCl ≤60 mL/min).67 mated by the MDRD formula).69 This study showed a direct
The randomized studies presented in this section have largely relationship between degree of renal impairment and the
excluded patients with severe renal impairment. In aggregate, risk of death, MI, refractory ischemia, and bleeding. Among
all demonstrate a stepwise increase in the incidence of death, patients with a GFR ≥58 mL·min−1·1.73 m−2, no significant
MI, and bleeding with increasing levels of kidney dysfunc- difference was seen between treatment groups in the primary
tion. However, the differential treatment effect of UFH versus composite outcome of death, MI, or refractory ischemia at 9
LMWH on bleeding is more difficult to ascertain because of days. However, at 30 days, the rate of the primary compos-
a lack of power caused by the small number of patients with ite outcome was significantly lower among patients with a
severe renal impairment.65,68 Of the trials discussed in this GFR <58 mL·min−1·1.73 m−2 with fondaparinux (HR, 0.81;
section, only the ExTRACT-TIMI 25 trial demonstrated an 95% CI, 0.69–0.96). Fondaparinux treatment was associated
association between treatment with enoxaparin and increased with lower rates of major bleeding events at 9 days among
bleeding risk with worsening renal function.67 A meta-­analysis patients with a GFR <58 mL·min−1·1.73 m−2 (HR, 0.42; 95%
of LMWH studies, including smaller randomized trials and CI, 0.32–0.56). In addition, in the subgroup of patients with
observational studies in ACS and venous thromboembolism CrCl <30 mL/min, the rates of major bleeding at 9 days were
Washam et al   Pharmacotherapy in CKD Patients With ACS   1137

significantly lower in the group receiving fondaparinux (2.4% in the rates of the primary composite ischemic end point of
versus 9.9%, P=0.001).75 Although the rates of major bleeding in-hospital death, Q-wave MI, and urgent target-vessel revas-
were lower with fondaparinux across all quartiles of estimated cularization (1.8% versus 0.8%, respectively; P=0.7).
GFR, the difference was most marked among patients with a Analyses from 2 randomized trials provide data on the asso-
GFR <58 mL·min−1·1.73 m−2.69 ciation of CKD with outcomes among patients with ACS receiv-
ing bivalirudin. In a substudy of the Acute Catheterization and
Direct Thrombin Inhibitors Urgent Intervention Triage Strategy (ACUITY) trial, Mehran
and colleagues82 showed that patients with CKD (defined as
Bivalirudin CrCl <60 mL/min) had worse 30-day and 1-year clinical out-
Bivalirudin, a bivalent direct thrombin inhibitor, has been well comes than those with normal renal function. Patients with
studied in patients undergoing PCI, including patients present- CrCl <30 mL/min were excluded from this trial. There were
ing with ACS. Current guidelines recommend bivalirudin as an no significant differences between bivalirudin monotherapy
anticoagulant option in STEMI patients undergoing primary and heparin plus a GP IIb/IIIa receptor antagonist in rates
PCI (Class I) and in UA/NSTEMI patients in whom an inva- of 30-day composite ischemia (11.1% versus 9.4%; P=0.27)
sive strategy is selected (Class I).26,42 Elimination of bivalirudin and net clinical adverse outcomes (16.1% versus 16.9%;
occurs through both proteolysis and renal clearance. In patients P=0.65). There was significantly less major bleeding (6.2%
with normal renal function (CrCl > 90mL/min) or mildly versus 9.8%; P=0.008) at 30 days but no significant differ-
impaired renal function (CrCl >60 mL/min), the pharmacoki- ence in 1-year composite ischemia (22.0% versus 18.9%;
netics of bivalirudin are linear, with an elimination half-life of P=0.10) or mortality (7.1% versus 7.3%; P=0.96). A substudy
25 minutes. The elimination half-life is increased to 34 to 57 of the Harmonizing Outcomes With Revascularization and
minutes in patients with moderate to severe renal impairment Stents in Acute Myocardial Infarction (HORIZONS-AMI)
(CrCl between 10 and 59 mL/min) and ≈3.5 hours in patients trial provided an assessment of the association of CKD with
with renal failure necessitating hemodialysis.76–78 For patients outcomes with bivalirudin use among patients undergoing pri-
with renal insufficiency, the product labeling recommends no mary PCI. Approximately 15% of patients enrolled had CrCl
reduction in the bolus dose for any degree of renal impair- of 30 to 60 mL/min, whereas 1.4% had CrCl of ≤30 mL/min.
ment, although the infusion dose of bivalirudin may need to This analysis showed patients with CKD had higher rates of
be reduced and anticoagulant status monitored in patients with clinical events at 30 days, including death and major bleed-
renal impairment (Table 1).28 Data from randomized trials and ing.83 Multivariable analysis identified baseline creatinine as
observational studies on the use of bivalirudin in ACS patients an independent predictor of death at 3 years (HR, 1.51; 95%
Downloaded from http://ahajournals.org by on January 29, 2019

with CKD are shown in Table 7. CI, 1.21–1.87; P<0.001). Patients with CKD (defined as CrCl
A meta-analysis79 of 3 randomized trials comparing bivali- <60 mL/min) randomized to bivalirudin monotherapy versus
rudin with UFH at the time of PCI (majority ACS) strati- heparin plus GP IIb/IIIa receptor antagonist had no significant
fied by CrCl >90 mL/min (n=1578), CrCl 90 to 60 mL/ difference in major bleeding (12.3% versus 15.3%; HR, 0.79;
min (n=2163), CrCl 59 to 30 mL/min (n=1255), and CrCl 95% CI, 0.50–1.25) or death (8.4% versus 7.9%; HR, 1.09;
<30 mL/min (n=39) showed an increasing risk of ischemic and 95% CI, 0.61–1.95) at 30 days.
bleeding events with increasing degrees of renal impairment. Taken collectively, the randomized controlled trial data on
The absolute benefit of bivalirudin on the ischemic and bleed- anticoagulation therapy in CKD patients presenting with ACS
ing composite end point increased with decreasing CrCl strata are limited by the underrepresentation of patients with stage
(2.2%, 5.8%, 7.7%, and 14.4%, respectively; Pinteraction=0.044). 4 and 5 CKD, which makes definitive conclusions about the
Similarly, the association of CKD with adverse outcomes for treatment effect of anticoagulant agents challenging. Although
patients undergoing PCI (43% ACS) randomized to bivalirudin a number of trials excluded patients with CrCl <30 mL/min,
and provisional GP IIb/IIIa inhibition or UFH and planned GP the ExTRACT-TIMI-25, OASIS-5, and HORIZONS-AMI
IIb/IIIa inhibition was examined in the Second Randomized trials did not.75,84,85 The association of treatment with enoxa-
Evaluation in PCI Linking Bivalirudin to Reduced Clinical parin and increased bleeding risk with worsening renal func-
Events (REPLACE-2) trial.80 Patients were grouped according tion observed in the ExTRACT-TIMI-25 trial, as well as
to CrCl ≥60 mL/min (n=4824) and CrCl <60 mL/min (n=886). the increased rates of major bleeding in patients with CrCl
Among patients with CrCl <60 mL/min, rates of 30-day isch- <30 mL/min with enoxaparin therapy observed in the OASIS-5
emic events (death, MI, or urgent revascularization) were 9.7% trial, suggests that caution is warranted when enoxaparin is
and 9.4% (P=0.870) in the bivalirudin and UFH and GP IIb/ used in this population.67,75
IIIa receptor antagonist groups, respectively, but there were
significantly lower rates of TIMI major or minor hemorrhage
Anti-Ischemic Therapies
(3.2% versus 7.1%; P=0.009) with bivalirudin. There was no
observed interaction between treatment, bleeding or ischemic β-Blockers
events, and renal function. Although these bivalirudin studies β-Blockers are recommended for all patients with ACS unless
excluded dialysis-dependent patients, a retrospective analysis contraindicated.86,87 Metoprolol, atenolol, and propranolol have
comparing bivalirudin with UFH (GP IIb/IIIa receptor antago- been studied in the setting of AMI, and carvedilol has been stud-
nists were excluded) in dialysis-dependent patients undergoing ied in the setting of AMI with left ventricular dysfunction.88–92
PCI showed no significant difference in the rates of in-hospital Atenolol is renally eliminated and requires dose adjustment in
major bleeding (3.4% versus 3.1%, respectively; P=0.9)81 or those with renal impairment. Dose adjustment is recommended
1138  Circulation  March 24, 2015

Table 7.  Summary of Bivalirudin Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
Chew et al 79
Meta-analysis The trials included patients undergoing Primary efficacy end Adverse ischemic and bleeding events increased
of 3 trials: the PCI receiving bivalirudin or UFH during point: composite of with worsening renal function. The ORs (95% CIs)
Bivalirudin PCI. Patients were stratified by CrCl* into death, MI, or urgent for reduction of the composite ischemic end point
Angioplasty Study, groups: CrCl >90 mL/min (n=1578), CrCl revascularization with bivalirudin in the 4 CrCl strata were 0.79
CACHET trial and 60–90 mL/min (n=2163), CrCl 30–59 (0.52–1.18), 0.73 (0.53–0.99), 0.77 (0.55-1.07),
REPLACE-1 mL/min (n=1255), and CrCl <30 mL/min and 0.81 (0.12–5.23), respectively.
(n=39). Patients with SCr >3 mg/dL were Bleeding (definition The ORs (95% CIs) for reduction in bleeding
excluded from the trials. The indication for varied by trial) events were 0.45 (0.21–0.96), 0.40 (0.09–1.86),
PCI was UA in ≈65% of patients in and 0.46 (0.30-0.70) for patients in the CrCl >90
this analysis. mL/min, CrCl 60–90 mL/min, and CrCl 30–59
mL/min groups, respectively. The absence of
bleeding events in the CrCl <30 mL/min bivalirudin
group precluded an estimate of relative benefit.
REPLACE-280 Subgroup analysis Patients undergoing PCI randomized Primary efficacy end A significant increase in adverse events was noted
of an RCT to bivalirudin and provisional GP IIb/IIIa point: 30-d composite in the group with renal insufficiency. In patients
inhibitor vs UFH and planned GP IIb/IIIa of death, MI, or urgent with CrCl <60 mL/min, the rates of the composite
inhibitor. Exclusion criteria included revascularization ischemic outcome in the bivalirudin and UFH plus
SCr >4 mg/dL. In this analysis, patients GP IIb/IIIa inhibitor groups were 7.4% and 6.7%
were grouped based on CrCl*: CrCl ≥60 (P=0.276), respectively.
mL/min (n=4824) and CrCl <60 mL/min Major bleeding The rates of protocol-defined major bleeding
(n=886). The indication for PCI was ACS were 5.1% in the bivalirudin group vs 7.1% in
in ≈43% of patients enrolled in this trial. the UFH plus GP IIb/IIIa inhibitor group (P=0.205),
respectively. In patients with CKD, the rate of TIMI
major bleeding events was significantly less in the
bivalirudin group (3.2% vs 7.1%; P=0.009).
Delhaye et al81 Retrospective Chronic dialysis-dependent patients Primary ischemic end The rates of the composite ischemic end point in
analysis of undergoing PCI receiving adjusted-dose point was the composite the bivalirudin and UFH groups were 1.8% and
single-center bivalirudin (n=267) or UFH (n=129). of in-hospital death, 0.8% (P=0.7), respectively.
registry Patients receiving GP IIb/IIIa inhibitor were Q-wave MI, or urgent TVR
Downloaded from http://ahajournals.org by on January 29, 2019

excluded. ACS was the indication for PCI In-hospital major bleed The rate of in-hospital major bleeding was
in 77% of patients receiving bivalirudin 3.4% vs 3.1% (P=0.9) in the bivalirudin and UFH
and 84% of those receiving UFH. groups, respectively.
ACUITY82 Subgroup analysis The ACUITY trial enrolled moderate- to Composite ischemic Patients with CKD had worse 30-d and 1-y
of an RCT high-risk ACS patients undergoing an end point of death, outcomes. At 30 d, in patients with CKD (CrCl
early invasive management strategy. MI, or unplanned <60 mL/min), the rates of the composite
Patients were randomized to 1 of 3 revascularization ischemic outcome in the bivalirudin alone vs
groups: a heparin (UFH or enoxaparin) Net clinical outcome heparin plus GP IIb/IIIa inhibitor groups were
plus a GP IIb/IIIa inhibitor, bivalirudin plus (ischemic composite or 11.1% vs 9.4%, respectively (RR, 1.18; 95%
a GP IIb/IIIa inhibitor, or bivalirudin with major bleeding) CI, 0.88-1.57). No significant difference was
provisional GP IIb/IIIa inhibitor use. Patients seen in the net clinical outcome at 30 d between
were grouped based on calculated CrCl*: treatment groups in those with CKD.
CrCl ≥60 mL/min (n=10 470) and CrCl Non-CABG-related Rates of 30-d major bleeding (non–CABG related)
<60 mL/min (n=2469). Although CrCl major bleeding were 6.2% vs 9.8% (RR, 0.64; 95% CI, 0.45–
<30 mL/min was an exclusion criterion, 0.89) favoring bivalirudin alone.
189 patients were enrolled.
HORIZONS-AMI83 Subgroup analysis Patients undergoing primary PCI for NACE (30 d): Major Patients with CKD (CrCl <60 mL/min) had higher
of an RCT STEMI randomized to bivalirudin or UFH bleeding (non–CABG rates of NACE and major bleeding (non–CABG related)
plus a GP IIb/IIIa inhibitor. Patients were related), or a composite at 30 d, 1 y, and 3 y. In patients with CKD, 30-d
grouped based on CrCl*: CrCl ≥60 mL/ of MACE including death, event rates and HRs (95% CIs) for patients receiving
min (n=2783) and CrCl <60 mL/min MI, stroke, or TVR for bivalirudin vs UFH plus a GP IIb/IIIa inhibitor were as
(n=554). There were 48 patients with ischemia follows: MACE, 12.7% vs 10.6% (1.22; 0.75–1.99);
CrCl <30 mL/min NACE 21.1% vs 21.6% (0.98; 0.68–1.41).
Non–CABG-related Rate of major bleeding (non-CABG) for bivalirudin
major bleeding (30 d) vs UFH plus a GP IIb/IIIa inhibitor was 12.3% vs
15.3% (HR, 0.79; 95% CI, 0.50–1.25).
ACS indicates acute coronary syndrome; ACUITY, Acute Catheterization and Urgent Intervention Triage Strategy; CABG, coronary artery bypass grafting; CACHET,
Comparison of Abciximab Complications With Hirulog for Ischemic Events Trial; CI, confidence interval; CKD, chronic kidney disease; CrCl, creatinine clearance; GP,
glycoprotein; HORIZONS-AMI, Harmonizing Outcomes With Revascularization and Stents in Acute Myocardial Infarction; HR, hazard ratio; MACE, major adverse cardiac
events; MI, myocardial infarction; NACE, net adverse clinical events; OR, odds ratio; PCI, percutaneous coronary intervention; RCT, randomized controlled trial; REPLACE,
Randomized Evaluation in PCI Linking Bivalirudin to Reduced Clinical Events; RR, relative risk; SCr, serum creatinine; TIMI, Thrombolysis in Myocardial Infarction; TVR,
target-vessel revascularization; UA, unstable angina; and UFH, unfractionated heparin.
*CrCl calculated by the Cockcroft-Gault formula.
Washam et al   Pharmacotherapy in CKD Patients With ACS   1139

with CrCl <35 mL/min (50 mg once daily for 15–35 mL/min ACS for patients with left ventricular systolic dysfunction at
and 25 mg once daily for CrCl <15 mL/min).93 Metoprolol, pro- the time of hospital discharge has become an important report-
pranolol, and carvedilol are all extensively hepatically metabo- able quality performance measure via which many hospitals
lized, with <5% of an oral dose excreted in the urine unchanged, are compared.101 Unfortunately, among patients with renal
so they do not require dose adjustments in renal impairment. dysfunction, these evidenced-based pharmacotherapies in the
Observational studies assessing CKD patients have analyzed hospital and at discharge are often significantly underused, par-
all β-blockers together. Data on the use of β-blockers for ACS ticularly for those with ESRD.3,19,21,102–107
patients with CKD are summarized in Table 8. The most common concerns with ACE inhibitors or ARBs
Limited data are available from randomized trials on include perceived worsening renal function and hyperkale-
the use of β-blockers in ACS in patients with CKD. A post mia.108 There is no absolute level of SCr that precludes the use
hoc analysis of pooled data from patients enrolled in the of these agents; however, if the SCr exceeds 2.5 mg/dL, cau-
Carvedilol Postinfarct Survival Control in Left Ventricular tion is warranted.109,110 In a review of 12 randomized controlled
Dysfunction (CAPRICORN) and Carvedilol Prospective trials of ACE inhibitor use in patients with renal dysfunction
Randomized, Cumulative Survival (COPERNICUS) trials (SCr >1.4 mg/dL), acute increases in SCr of up to 30% that
assessed carvedilol therapy in MI patients with left ventricu- stabilized within the first 2 months of therapy initiation were
lar dysfunction (CAPRICORN) and in chronic systolic heart associated with a 55% to 75% risk reduction in renal disease
failure patients with CKD (COPERNICUS).94 In the group of progression compared with those with normal renal func-
patients with CKD (defined as eGFR ≤60 mL·min−1·1.73 m−2), tion.111 Practically, the use of ACE inhibitors or ARBs can be
treatment with carvedilol was associated with a reduction in considered in patients with CKD as long as the SCr does not
all-cause mortality (HR, 0.76; 95% CI, 0.63–0.93) and cardio- increase beyond this point and the serum potassium remains
vascular mortality (HR, 0.77; 95% CI, 0.62–0.94). However, <5.5 mEq/L.112 For patients with ESRD, the administration of
a sensitivity analysis among the subgroup of patients with an these medications can be problematic. The use of ACE inhibi-
eGFR <45 mL·min−1·1.73 m−2 (n=1116) failed to show a signif- tors or ARBs in patients undergoing chronic dialysis has been
icant benefit for carvedilol therapy with regard to the primary or associated with an increased risk of hyperkalemia, although
secondary outcomes. Several observational cohort studies have study results have been variable.113–115
evaluated whether β-blockers were beneficial among patients Most of the large randomized controlled trials evaluat-
with various degrees of renal impairment. These observational ing the effectiveness of ACE inhibitors or ARBs in post-
studies have found that the benefit of discharge β-blocker use MI patients with left ventricular dysfunction have excluded
was preserved across all degrees of renal dysfunction.2,32,38,95 patients with ESRD, with SCr cut offs ranging from 2 to
Downloaded from http://ahajournals.org by on January 29, 2019

One of the largest observational studies evaluated Medicare 3.4 mg/dL. Nonetheless, the ability of these agents to prevent
beneficiaries with AMI included in the ESRD program as part ventricular dilation and to significantly improve mortality for
of the Cooperative Cardiovascular Project (CCP).19 This study patients with compromised cardiac function should not be
identified a cohort of 145 765 patients with AMI, among whom underestimated. Although randomized trials of ACE inhibi-
1025 had stage 5D CKD. They found that although β-blocker tor therapy in ACS have systematically excluded patients with
use did not reduce mortality to as great an extent among those ESRD, the Fosinopril in Dialysis (FOSIDIAL) study was
receiving dialysis as among those without ESRD, the benefit undertaken in chronic ESRD patients to assess the impact of
was still significant. β-Blocker use was associated with a 40% fosinopril therapy on cardiovascular events.116 No significant
relative reduction in mortality among those undergoing dialy- benefit was seen with fosinopril in the intention-to-treat analy-
sis and a 56% relative reduction among those without ESRD sis for the composite of cardiovascular events (RR, 0.93; 95%
(P<0.001 for both groups).19 CI, 0.68–1.26), although a per protocol analysis suggested a
Collectively, the data from randomized and observational trend toward benefit for fosinopril (RR, 0.79; 95% CI, 0.59–
studies support the routine use of β-blocker therapy in CKD 1.1). Subgroup analyses from randomized trials and observa-
patients presenting with ACS when no contraindications are tional studies have suggested that among patients with ACS
present. with reduced left ventricular ejection fraction, the use of ACE
inhibitors or ARBs may be more beneficial when renal insuf-
Angiotensin Blockade ficiency coexists.103,117–122 Data on the use of ACE inhibitors or
For patients with ACS, angiotensin-converting enzyme (ACE) ARBs in ACS patients with CKD are summarized in Table 9.
inhibitors are recommended by current guidelines to be initiated In summary, the data on the use of ACE inhibitors in patients
and continued indefinitely in all patients with a left ventricular with post-MI left ventricular dysfunction and CKD consis-
ejection fraction <40% and for those with hypertension, diabe- tently show improved outcomes with ACE inhibitor therapy.
tes mellitus, and CKD unless contraindicated.73,87 For patients Caution is warranted at the time of initiation, and monitoring
who are considered intolerant to ACE inhibitors, angiotensin of serum creatinine and potassium is required.
receptor blockers (ARBs) can be considered as an alterna-
tive.73,87 These Class I recommendations are based on data from Aldosterone Receptor Antagonists
several large randomized trials and meta-analyses documenting According to the American College of Cardiology/American
significant reductions in mortality among patients with ACS, in Heart Association guidelines, the use of aldosterone antagonists
which the greatest benefit was demonstrated when angiotensin such as spironolactone and eplerenone have a Class IA recom-
blockade was administered within the first 24 hours after an mendation for post-MI patients who are receiving therapeutic
MI.96–100 Additionally, receipt of an ACE inhibitor or ARB after doses of an ACE inhibitor and a β-blocker, have a left ventricular
1140  Circulation  March 24, 2015

Table 8.  Summary of β-Blocker Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
Wali et al
94
Post hoc analysis of 4217 patients with either post-MI left All-cause mortality No statistically significant interactions were
pooled data from 2 ventricular dysfunction (CAPRICORN) (primary) observed between the randomized treatment
RCTs (CAPRICORN and or severe chronic HF (COPERNICUS) Cardiovascular and the study type for each clinical outcome.
COPERNICUS) randomized to carvedilol or placebo; 46% mortality, HF mortality Adjusted HRs (95% CI) for carvedilol treatment
of patients were from the CAPRICORN trial. were as follows: All-cause mortality: eGFR >60:
Patients categorized based on eGFR*: eGFR 0.59 (0.43–0.81); eGFR ≤60: 0.78 (0.64-0.95).
>60 mL/min/1.73 m2 (n=1,651), eGFR ≤60 In the subgroup with eGFR <45, HR was 0.94
(n=2,566). Among those with eGFR ≤60, (95% CI, 0.72–1.23). Cardiovascular mortality:
1,116 had an eGFR <45 mL/min/1.73 m2 eGFR >60: 0.59 (0.42-0.82); eGFR ≤60: 0.77
(0.62–0.94).
HF mortality: eGFR >60: 0.58 (0.34-0.99);
eGFR ≤60: 0.68 (0.52–0.88); in the subgroup
with eGFR <45: 0.86 (0.61–1.21).
Yan et al38 Observational, 3510 patients hospitalized for ACS with One-year survival β-Blockers were associated with improved 1-y
prospective normal renal function (CrCl† ≥90 mL/min; survival to a similar extent among those with
cohort study n=1152), mild renal dysfunction (CrCl 60–89 normal and impaired renal function. Discharge
mL/min; n=1253), moderate renal dysfunction β-blocker use–adjusted OR, 0.76 (95% CI,
(CrCl 30–59 mL/min; n=944), and severe 0.56–1.02), P=0.07; P=0.37 for heterogeneity
renal dysfunction (CrCl <30 mL/min; n=161) across CrCl <60 vs ≥60 mL/min.
Wright et al2 Observational, retro­ 3106 patients with AMI with no renal disease Short- and long-term β-Blockers were associated with improved
spective cohort study (n=1320), mild chronic renal insufficiency survival stratified postdischarge survival across the spectrum
(CrCl >50 but ≤75 mL/min; n=860), moderate by CrCl of renal failure (adjusted HR, 0.7; 95% CI,
renal dysfunction (CrCl >35 but ≤50 mL/min; 0.6–0.9); P<0.001).
n=491), severe renal insufficiency (CrCl†
<35 mL/min; n=391), or ESRD (n=44)
Bae et al95 Observational, 13 901 consecutive patients with AMI Short and long-term Use of β-blockers, ACE inhibitors, and statins
retrospective analysis divided into strata based on eGFR* ≥90 MACE (analyzed together) was associated with decreased
of KAMIR cohort (n=3491; reference), ≥60 to <90 (n=5791, risk of short- and long-term MACE. HRs (95% CIs)
group 2), ≥30 to <60 (n=2609, group 3), improved across the spectrum of renal impairment
Downloaded from http://ahajournals.org by on January 29, 2019

≥15 to <30 (n=439, group 4), and <15 when discharge medication use was added to
(n=306, group 5) mL·min−1·1.73 m−2 model that included age, Killip class >1, diabetes
mellitus and hypertension, hs-CRP, and PCI. Group
2: 0.93 (0.79–1.09) vs 0.97 (0.82–1.14); group
3: 1.58 (1.32-1.90) vs 1.73 (1.44–2.07); group 4:
2.12 (1.63–2.75) vs 2.26 (1.74–2.93); and group 5:
2.50 (1.89–3.29) vs 2.69 (2.04–3.56). Additionally,
1-mo and 1-y MACE were improved in a stepwise
fashion with 3 medications vs 2 medications vs 1
medication vs no medications.
Keough-Ryan Observational cohort 5549 consecutive patients with ACS. Death, secondary Discharge β-blockers were associated with
et al32 Renal function classified as >80 (n=1430), outcomes length significant reduction in mortality (HR, 0.91; 95%
60–80 (n=2018), 30–59 (n=1795), and of stay, surgical CI, 0.855-0.97), with no interaction with degree
<30 (n=306) mL·min−1·1.73 m−2 * interventions of renal impairment (P>0.10).
Berger et al19 Observational, AMI patients: 145 740 patients without Mortality (30 d) β-Blocker therapy was associated with a 40%
retrospective analysis ESRD and 1025 patients with ESRD relative reduction in mortality in those receiving
of CCP and USRDS receiving dialysis dialysis (P<0.001) and a 56% relative reduction
among those without ESRD (P<0.001).
McCullough Observational, 1724 patients with STEMI. Renal function In-hospital mortality Adjusted RR reduction for the combination of
et al21 prospective was classified by quartile corrected in-hospital aspirin and β-blocker was 80%,
cohort study creatinine clearance‡ >81.5 mL/min 74.9%, 69%, 64.3%, and 77.9% across the
(n=524), 63.1 to ≤81.5 (n=421), 46.2 to quartiles of corrected CrCl, respectively.
≤63.1 (n=421), ≤46.2 not undergoing
dialysis (n=310), and chronic dialysis (n=47).
ACE indicates angiotensin-converting enzyme; ACS, acute coronary syndrome; AMI, acute myocardial infarction; CAPRICORN, Carvedilol Postinfarct Survival Control
in Left Ventricular Dysfunction; CCP, Cooperative Cardiovascular Project; CI, confidence interval; CKD, chronic kidney disease; COPERNICUS, Carvedilol Prospective
Randomized, Cumulative Survival; CrCl, creatinine clearance; eGFR, estimated glomerular filtration rate; ESRD, end-stage renal disease; HF, heart failure; HR, hazard
ratio; hs-CRP, high-sensitivity C-reactive protein; KAMIR, Korean Acute Myocardial Infarction Registry; MACE, major adverse cardiovascular events; MI, myocardial
infarction; OR, odds ratio; PCI, percutaneous coronary intervention; RCT, randomized controlled trial; RR, relative risk; STEMI, ST-segment–elevation myocardial
infarction; and USRDS, United States Renal Data System.
*Glomerular filtration rate calculated by the modified Modification of Diet in Renal Disease equation.
†CrCl calculated via Cockcroft-Gault equation.
‡Corrected CrCl calculated as (140−age)/serum creatinine, multiplied by 0.85 if female.
Washam et al   Pharmacotherapy in CKD Patients With ACS   1141

Table 9.  Summary of ACE Inhibitor Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
Frances et al, Observational, 20 902 Medicare beneficiaries admitted All-cause In the multivariate analysis, the HR (95% CI) for
2000118 retrospective cohort for MI with LVEF <40%; 19 320 with SCr mortality (1 y) ACE inhibitor therapy at hospital on discharge
of the CCP database ≤3 mg/dL and 1582 with SCr >3 mg/dL on mortality at 1 y was 0.84 (­0.77–0.92) in
those with SCr ≤3 mg/dL and 0.63 (0.48–0.84)
in those with SCr >3 mg/dL.
Shlipak et al, Observational, 20 902 Medicare beneficiaries admitted All-cause In the multivariate analysis, ACE inhibitor
2001122 retrospective for MI with LVEF <40%. SCr ≥2 mg/dL mortality (1 y) exposure was associated with a 23% increase
analysis of the CCP was seen in 4133 patients. in 1-y survival for patients with SCr ≥2 mg/dL
database (HR, 0.77; 95% CI, 0.66–0.90).
Berger et al, Observational, 145 740 patients without ESRD and 1025 All-cause Compared with ESRD patients not receiving
200319 retrospective cohort patients with ESRD admitted for MI using mortality (30 d) an ACE inhibitor, those with ESRD receiving
using ESRD and CCP peritoneal dialysis or hemodialysis before an ACE inhibitor exhibited a 48% lower 30-d
database admission mortality (P<0.001).
CATS119 Post hoc analysis 298 patients with a first anterior wall MI Change in GFR* Patients receiving captopril had an annual
of an RCT from baseline over decline of only 0.5 mL/min in GFR vs 5.5 mL/min
a 12-mo period in the placebo group (P<0.05). Patients with
compromised renal function at baseline (<81
mL/min) had the greatest improvement in GFR.
SAVE121 Post hoc analysis 2183 patients with LVEF ≤40% post-MI All-cause and The relative risk reduction for all-cause
of an RCT with SCr <2.5 mg/dL. Patients were cardiovascular vcardiovascular mortality/morbidity, the
grouped by eGFR*: eGFR >60 (n=1464) mortality and morbidity relative risk reduction was higher for those
and eGFR <60 mL·min−1·1.73 m−2 stratified treated with captopril with CKD than for those
(n=719). by GFR* (4 y) without CKD (31% vs 20%, respectively;
P=0.29 for interaction).
Krause et al, Observational, 1342 Medicare beneficiaries post-MI with 300–400-d all-cause In the adjusted analysis, patients receiving
2004120 retrospective cohort mild (GFR 60–89), moderate (GFR 30–59), mortality aspirin, β-blocker, and ACE inhibitor exhibited
of the CCP and severe (GFR 15–29 mL·min−1·1.73 the following reductions in mortality based on
m−2) renal dysfunction† renal function: mild, HR 0.54 (95% CI, 0.26-
1.12); moderate, HR 0.50 (95% CI, 0.28–0.88);
Downloaded from http://ahajournals.org by on January 29, 2019

and severe, HR 0.35 (95% CI, 0.09–1.42).


Reddan et al, Post hoc analysis of 13 707 patients with CKD post-MI grouped Association between The interaction between use of ACE inhibitors
2005103 the SYMPHONY trials based on CrCl: ≥90 (n=6840), 60–89 CrCl and 90-d all-cause and CrCl was significantly associated with
(n=5909), and 30–59 mL/min (n=955) mortality‡ improved outcomes, with the greatest
benefit in seen in patients with CrCl of 30–59
mL·min−1·1.73 m−2 (P=0.0005 for interaction).
ACE indicates angiotensin-converting enzyme; ACS, acute coronary syndrome; CATS, Captopril and Thrombolysis Study; CKD, chronic kidney disease; CCP, Cooperative
Cardiovascular Project; CI, confidence interval; CrCl, creatinine clearance; eGFR, estimated glomerular filtration rate; ESRD, end-stage renal disease; GFR, glomerular
filtration rate; HR, hazard ratio; LVEF, left ventricular ejection fraction; MI, myocardial infarction; RCT, randomized controlled trial; SAVE, Survival And Ventricular Enlargement
study; SCr, serum creatinine; and SYMPHONY, Sibrafiban Versus Aspirin to Yield Maximum Protection From Ischemic Heart Events Post-Acute Coronary Syndromes.
*GFR calculated by the modified Modification of Diet in Renal Disease equation.
†GFR calculated via Cockcroft-Gault equation.
‡CrCl calculated by modified Modification of Diet in Renal Disease equation.

ejection fraction <40%, and have either diabetes mellitus or heart publication of the Randomized Aldactone Evaluation Study
failure.87 These recommendations are primarily derived from (RALES), concerns have been raised regarding the increased
the Eplerenone Post-Acute Myocardial Infarction Heart Failure risk of life-threatening hyperkalemia attributable to aldoste-
Efficacy and Survival (EPHESUS) trial, which demonstrated a rone antagonists, particularly when combined with angiotensin
15% all-cause mortality risk reduction (P=0.008) and a 13% blockade.124 Therefore, the American College of Cardiology/
reduction in cardiovascular-related mortality or cardiovascular American Heart Association guidelines recommend against
hospitalizations (P=0.002) among post-MI patients with a left using aldosterone blockade if significant renal dysfunction (SCr
ventricular ejection fraction ≤40% who received eplerenone 25 >2.5 mg/dL in men and >2.0 mg/dL in women) or hyperkale-
to 50 mg/d in addition to background heart failure therapy with mia (serum potassium >5.0 mEq/L) coexists.87
an ACE inhibitor or ARB and β-blocker.123 However, important A post hoc analysis of the EPHESUS trial evaluated serial
exclusion criteria for the EPHESUS trial were SCr >2.5 mg/dL changes in eGFR related to eplerenone use in patients after
or a serum potassium >5.0 mEq/L. Serious hyperkalemia (≥6.0 ACS. In this analysis, Rossignol et al125 found that 5792 patients
mEq/L) occurred in 5.5% of patients receiving eplerenone com- assigned to eplerenone 25 to 50 mg/d had a significant decline
pared with 3.9% in the placebo group (P=0.002). For patients in their eGFR, with a mean adjusted difference of −1.4±0.3
with CrCl <50 mL/min, the incidence of serious hyperkalemia mL·min−1·1.73 m−2 compared with placebo (P<0.0001). This
increased to 10.1% among those receiving eplerenone com- effect occurred within the first month and persisted throughout
pared with 5.9% in the placebo group (P=0.006). Since the the 24-month follow-up. Patients receiving eplerenone had
1142  Circulation  March 24, 2015

Table 10.  Summary of Statin Studies in Patients With ACS and CKD
Study Study Design Population End Points Results
Lim et al 132
Observational, 12 865 AMI patients; 3256 patients with Death and Adjusted for covariates, HR (95% CI) 2.2
retrospective analysis AMI and renal insufficiency (eGFR<60 complications during (1.696–2.881), P<0.001 in renal dysfunction
of KAMIR cohort mL/min)* were included; 2218 were taking hospital course group with no statin therapy vs other groups.
statins and 1038 were not For eGFR <30 but ≥15 mL/min and no statin
therapy: HR, 2.0; 95% CI, 1.2–3.5; P=0.008.
For eGFR <15 mL/min: HR, 2.3; 95% CI, 1.1–
4.8; P=0.036 compared with statin therapy.
CARE135 Post hoc analysis 1711 participants with CrCl† ≤75 mL/min Death of coronary Adjusted for covariates, HR (95% CI) with
of an RCT with AMI 3–20 mo before randomization disease or symptomatic pravastatin 0.72 (0.55–0.95); P=0.02
randomized to pravastatin or placebo nonfatal MI confirmed compared with placebo.
by CK
Keough-Ryan Observational cohort 5549 consecutive patients with ACS. Death, secondary Discharge lipid-lowering therapy was
et al32 Renal function classified as >80 (n=1430), outcomes length associated with significant reduction in
60–80 (n=2018), 30–59 (n=1795), or <30 of stay, surgical mortality (HR, 0.835; 95% CI, 0.783–0.890)
(n=306) mL·min−1·1.73 m−2‡ interventions with no significant interaction with degree of
renal impairment (P>0.10).
Szummer Observational analysis 42 814 survivors of MI without statin therapy Mortality at 1y Statin therapy was associated with a
et al133 of the SWEDEHEART on admission classified into 5 renal function 37% reduction in risk of death (adjusted
registry stages according to eGFR‡: ≥90 (n=9935), HR, 0.63; 95% CI, 0.58–0.69; P<0.001).
60–89 (n=20 135), 30–59 (n=11 103), After adjustment, there was a significant
15–29 (n=1273), and <15 mL·min−1·1.73 interaction between statin therapy and
m−2 or undergoing dialysis (n=368) renal function stage (P<0.001). Benefit was
significant in all groups except those with
eGFR <15 mL·min−1·1.73 m−2 or undergoing
dialysis Adjusted HRs (95% CI) across the
groups were as follows: 0.53 (0.41–0.67),
0.60 (0.52–0.68), 0.67 (0.60–0.76), 0.72
(0.56–0.94), and 1.13 (0.76–1.67) in
≥90, 60–89, 30–59, 15–29, and <15
Downloaded from http://ahajournals.org by on January 29, 2019

mL·min−1·1.73 m−2/dialysis, respectively.


Shibui et al134 Observational cohort 501 patients presenting with ACS who Composite end point The composite end point occurred in 16.2%
underwent successful PCI; 324 patients of cardiac death or of CKD patients treated with statins compared
(64.7%) had CKD based on eGFR <60 readmissions for ACS with 26.3% of CKD patients not treated with
mL·min−1·1.73 m−2. Statin therapy was over a mean follow-up statin therapy (HR, 0.58; 95% CI, 0.34–0.98;
used in 34.3% of patients with CKD. of 5.2 y P=0.039).
ACS indicates acute coronary syndrome; AMI, acute myocardial infarction; CARE, Cholesterol and Recurrent Events trial; CI, confidence interval; CK, creatine kinase;
CKD, chronic kidney disease; CrCl, creatinine clearance; eGFR, estimated glomerular filtration rate; HR, hazard ratio; KAMIR, Korean Acute Myocardial Infarction Registry;
MI, myocardial infarction; PCI, percutaneous coronary intervention; RCT, randomized controlled trial; and SWEDEHEART, Swedish Web System for Enhancement and
Development of Evidence-Based Care in Heart Disease Evaluated According to Recommended Therapies.
*Glomerular filtration rate calculated via Cockcroft-Gault equation.
†CrCl calculated by modified Modification of Diet in Renal Disease equation.
‡Glomerular filtration rate calculated by the modified Modification of Diet in Renal Disease equation.

a 1.15-fold increased odds of experiencing a >20% decline CKD. Furthermore, patients with SCr >2 mg/dL were excluded
in eGFR within the first month compared with placebo from the original ACS randomized trials of early initiation of
(P=0.017).125 However, an eGFR ≤60 mL·min−1·1.73 m−2 was statins.126,127 However, in primary prevention, randomized tri-
not associated with early worsening renal function, and there als of statin therapy in patients with CKD have yielded mixed
was no interaction between worsening renal function and the results.128–131 Early studies found no benefit of statin therapy
favorable effects of eplerenone on cardiovascular death or among patients with CKD (mostly dialysis dependent),129–131
hospitalization (P=0.77 for interaction) and hospitalization which led to speculation that in ESRD, there may be a more
for heart failure (P=0.82 for interaction).125 advanced atherosclerotic state that leads to more sudden deaths
Statins caused by arrhythmias than among patients without ESRD.
A large body of literature supports of the use of statins after However, more recently, the largest of these studies, the Study
an ACS to reduce the risk of death or vascular events. Current of Heart and Renal Protection (SHARP), included patients
guidelines recommend statins regardless of baseline low-den- with CKD both on dialysis and not on dialysis and showed
sity lipoprotein level in all ACS patients in the absence of con- a benefit of simvastatin plus ezetimibe therapy on the risk of
traindications.26,27 With regard to CKD patients, those receiving the primary composite end point of first major atherosclerotic
chronic dialysis in particular, the use of statins has been more event (RR, 0.83; 95% CI, 0.74–0.94; P=0.0021), although no
controversial. No randomized controlled trials have assessed statistically significant difference was observed for the risk of
the safety and efficacy of statins in patients with ACS and vascular death (RR, 0.93; 95% CI, 0.80–1.07).128 Although
Washam et al   Pharmacotherapy in CKD Patients With ACS   1143

Table 11.  Summary of Evidence for Pharmacotherapy for ACS Patients With CKD*
Medication Class Summary
Aspirin Available data suggest aspirin should be used in patients with CKD presenting with ACS
Fibrinolytic Available data suggest fibrinolytic therapy should be considered as a treatment strategy in STEMI patients with CKD
presenting within 12 h of symptom onset when primary PCI is not available. Observed rates of ICH were higher in CKD
patients receiving fibrinolytics than in non-CKD patients
Oral P2Y12 receptor antagonist Available data suggest these agents should be considered in CKD patients presenting with ACS. Data from RCTs of newer
agents (prasugrel and ticagrelor) suggest these agents should be considered in CKD patients not requiring dialysis.
Glycoprotein IIb/IIIa receptor antagonist Available data suggest glycoprotein IIb/IIIa receptor antagonist therapy, within the context of labeled dosing modifications
and exclusions for each agent, can be considered as a treatment strategy in CKD patients presenting with ACS. The data
also suggest an increased rate of bleeding in patients with CKD.
Anticoagulant Available data suggest anticoagulant therapy should be considered in CKD patients presenting with ACS. The data support
consideration for fondaparinux and bivalirudin as strategies with lower rates of bleeding in patients with stage 3 and 4
CKD (relatively few patients with stage 4 CKD were included in the randomized trials evaluating these agents). Relevant
labeled dosing modifications and contraindications should be considered for each agent.
β-Blocker Available data suggest β-blocker therapy should be considered in CKD patients presenting with ACS who do not have a
contraindication to β-blocker therapy.
ACE inhibitor/ARB Available data suggest ACE inhibitor or ARB therapy should be considered in CKD patients presenting with ACS and LV
dysfunction. Potassium and SCr should be monitored closely.
Aldosterone blocker Limited available data suggest aldosterone blocker therapy should be considered for CKD patients with post-MI LV
dysfunction (with either diabetes mellitus or heart failure signs or symptoms) and baseline SCr ≤2.5 mg/dL and serum
potassium <5.0 mmol/L. Serum potassium should be monitored closely.
Statin Available data suggest statin therapy should be considered in CKD patients presenting with ACS.
ACE indicates angiotensin-converting enzyme; ACS, acute coronary syndrome; ARB, angiotensin receptor blocker; CKD, chronic kidney disease; ICH, intracranial
hemorrhage; LV, left ventricular; MI, myocardial infarction; PCI, percutaneous coronary intervention; RCT, randomized controlled trial; SCr, serum creatinine; and STEMI,
ST-segment–elevation myocardial infarction.
*A detailed discussion and all references can be found in the text of each respective drug section.

statin trials in ACS have largely excluded patients with CKD, for muscle-related side effects associated with statin therapy,136
Downloaded from http://ahajournals.org by on January 29, 2019

observational studies have evaluated the safety and efficacy of although randomized trials evaluating moderate-intensity statin
statins in these patients.32,132–134 The data on the use of statins therapy in patients with advanced CKD (including ESRD)
in ACS patients with CKD are summarized in Table 10. The without ACS have not supported this observation.128,129,131
Korea Acute Myocardial Infarction Registry (KAMIR) study
was observational and evaluated 12 865 patients with MI, Summary/Future Directions
3256 of whom had renal dysfunction (defined as eGFR <60 In patients presenting with ACS, declining renal function has
mL·min−1·1.73 m−2).132 The study found that patients with renal been associated with increased risk for adverse clinical outcomes,
dysfunction not taking statins were at significantly increased including death, MI, and bleeding events. In spite of the high risk
risk of in-hospital and 1-month major adverse cardiovascular of adverse events in this population, CKD patients have largely
events and cardiac death at 1 year. These results were also been excluded from or underrepresented in randomized controlled
consistent among those with severe renal insufficiency (eGFR trials in patients presenting with ACS. This presents a challeng-
<30 but ≥15 mL·min−1·1.73 m−2 and eGFR <15 mL·min−1·1.73 ing situation for clinicians to make evidence-based medication
m−2).132 Although the Cholesterol and Recurrent Events choices, as well as for understanding the risk and benefits of dif-
(CARE) study excluded participants with >2+ proteinuria or ferent therapy combinations. Taken collectively, the available data
SCr >1.5 times the upper limit of normal, subgroup analysis of suggest that patients with CKD benefit from the evidence-based
those with mild renal insufficiency (CrCl ≤75 mL/min) found medications routinely used in all patients presenting with ACS
that pravastatin reduced the risk of death of coronary disease (Table 11); however, important considerations are necessary to
or symptomatic nonfatal MI by 28% (adjusted HR, 0.72; 95% provide the greatest benefit while limiting the chance for harm.
CI, 0.55–0.95; P=0.02) among patients with AMI between 3 These would include careful assessment of renal function, use of a
and 20 months before randomization (Table 10).135 The analy- validated equation for dose adjustment of medications, avoidance
sis also found that adverse events were infrequent among those of medications that are contraindicated in patients with stage 4 and
with chronic renal insufficiency, with no significant differences 5 CKD, and avoidance or limiting of the use of emerging medica-
in frequency compared with placebo.135 tions that have not been formally studied in patients with CKD.
Taken together, these data show a consistent benefit with Currently, the US Food and Drug Administration provides
regard to a reduction in cardiovascular events with statin ther- guidance and recommendations for the pharmaceutical indus-
apy in CKD patients who present with ACS and support the try on the design and conduct of pharmacokinetic studies in
routine use of statins in this population. However, the data are patients with impaired renal function.137 However, moving for-
somewhat limited by the lack of information on statin dose, ward, inclusion and better representation of patients with CKD
as well as medication side effects. These are important con- in randomized clinical trials will be necessary to accurately
siderations, because patients with CKD may be at higher risk assess the risks and benefits of medications in this population.
1144  Circulation  March 24, 2015

Disclosures

Writing Group Disclosures


Speakers’
Writing Group Other Research Bureau/ Expert Ownership Consultant/Advisory
Member Employment Research Grant Support Honoraria Witness Interest Board Other
Jeffrey B. Duke Heart Center None None None None None None None
Washam
L. Kristin Duke University Amylin*; Bristol Myers- None None None None Amgen†; American
Newby Medical Center Squibb*; Duke†; AstraZeneca*; Heart Journal*;
Eli Lilly & Co*; Cubist*; Daiichi Society of Chest
GlaxoSmithKline†; Sankyo*; Pain Centers*;
Johnson & Johnson*; Genentech*; JAHA*
Merck & Co†; NIH†; GlaxoSmithKline*;
Regado Biosciences* Novartis*
Amber L. University of Maryland None None None None None None None
Beitelshees
Mauricio G. University of Miami None None None None None Daiichi-Sankyo†; None
Cohen Miller School of AstraZeneca†;
Medicine The Medicines
Company*
Timothy D. Cedars-Sinai Heart None None None None None Daiichi Sankyo*; None
Henry Institute Eli Lilly*; The
Medicines
Company*
Charles A. Hennepin Healthcare AHRQ DEcIDE Network†; Affymax*; Amgen†; CardioRenal None Boston Abbott*; Affymax*; None
Herzog System, Inc NIH/NIDDK†; NHLBI† Gilead†; Johnson & Society of Scientific*; Amgen†;
Johnson† America & NKF Cambridge CorMedix*;
of Arizona*; Heart*; FibroGen*;
Greenfield Johnson & Fresenius*; Keryx*;
Downloaded from http://ahajournals.org by on January 29, 2019

Health Johnson*; GlaxoSmithKline*;


System*; Merck* AbbVie*
Keryx*
Navin K. Kapur Tufts Medical Center None None None None None None None
Jessica L. Brigham & Women’s Daiichi Sankyo†; None None None None Bayer*; Janssen*; None
Mega Hospital AstraZeneca† Portola*
Venu Menon Cleveland Clinic AstraZeneca† None None None None None None
Robert L. University of Colorado None None None None None None None
Page II School of Pharmacy
This table represents the relationships of writing group members that may be perceived as actual or reasonably perceived conflicts of interest as reported on the
Disclosure Questionnaire, which all members of the writing group are required to complete and submit. A relationship is considered to be “significant” if (a) the person
receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the
entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.
*Modest.
†Significant.
Washam et al   Pharmacotherapy in CKD Patients With ACS   1145

Reviewer Disclosures
Other Research Speakers’ Bureau/ Ownership Consultant/
Reviewer Employment Research Grant Support Honoraria Expert Witness Interest Advisory Board Other
Sripal Bangalore New York University NHLBI (PI of the None None None None None None
ISCHEMIA CKD
trial)†
Gregory Barsness Mayo Clinic None None None None None None None
David Charytan Brigham and Women’s NIH† Medtronic† None Fresenius† None Keryx*; None
Hospital Medtronic*
Peter McCullough Baylor University Medical None None None None None None None
Center, Baylor Heart and
Vascular Institute, Baylor
Jack and Jane Hamilton
Heart and Vascular Hospital,
Dallas, TX, The Heart
Hospital, Plano, TX
This table represents the relationships of reviewers that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure
Questionnaire, which all reviewers are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during
any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more
of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.
*Modest.
†Significant.

References 12. Sosnov J, Lessard D, Goldberg RJ, Yarzebski J, Gore JM. Differential
symptoms of acute myocardial infarction in patients with kidney disease:
1. Fox CS, Muntner P, Chen AY, Alexander KP, Roe MT, Cannon CP, Saucedo
a community-wide perspective. Am J Kidney Dis. 2006;47:378–384. doi:
JF, Kontos MC, Wiviott SD; Acute Coronary Treatment and Intervention
10.1053/j.ajkd.2005.11.017.
Outcomes Network Registry. Use of evidence-based therapies in short-
13. Szummer K, Lundman P, Jacobson SH, Schön S, Lindbäck J, Stenestrand
term outcomes of ST-segment elevation myocardial infarction and non–ST-
U, Wallentin L, Jernberg T; SWEDEHEART. Influence of renal func-
segment elevation myocardial infarction in patients with chronic kidney
tion on the effects of early revascularization in non–ST-elevation myo-
disease: a report from the National Cardiovascular Data Acute Coronary
cardial infarction: data from the Swedish Web-System for Enhancement
Treatment and Intervention Outcomes Network registry. Circulation.
Downloaded from http://ahajournals.org by on January 29, 2019

2010;121:357–365. doi: 10.1161/CIRCULATIONAHA.109.865352. and Development of Evidence-Based Care in Heart Disease Evaluated
2. Wright RS, Reeder GS, Herzog CA, Albright RC, Williams BA, Dvorak According to Recommended Therapies (SWEDEHEART). Circulation.
DL, Miller WL, Murphy JG, Kopecky SL, Jaffe AS. Acute myocardial 2009;120:851–858. doi: 10.1161/CIRCULATIONAHA.108.838169.
infarction and renal dysfunction: a high-risk combination. Ann Intern 14. Shroff GR, Frederick PD, Herzog CA. Renal failure and acute myocardial
Med. 2002;137:563–570. infarction: clinical characteristics in patients with advanced chronic kid-
3. Shlipak MG, Heidenreich PA, Noguchi H, Chertow GM, Browner WS, ney disease, on dialysis, and without chronic kidney disease: a collabora-
McClellan MB. Association of renal insufficiency with treatment and tive project of the United States Renal Data System/National Institutes of
outcomes after myocardial infarction in elderly patients. Ann Intern Med. Health and the National Registry of Myocardial Infarction. Am Heart J.
2002;137:555–562. 2012;163:399–406. doi: 10.1016/j.ahj.2011.12.002.
4. Moscucci M, Fox KA, Cannon CP, Klein W, López-Sendón J, Montalescot 15. Szummer K, Lundman P, Jacobson SH, Schön S, Lindbäck J, Stenestrand
G, White K, Goldberg RJ. Predictors of major bleeding in acute coronary U, Wallentin L, Jernberg T; SWEDEHEART. Relation between renal
syndromes: the Global Registry of Acute Coronary Events (GRACE). Eur function, presentation, use of therapies and in-hospital complications in
Heart J. 2003;24:1815–1823. acute coronary syndrome: data from the SWEDEHEART register. J Intern
5. Coca SG, Krumholz HM, Garg AX, Parikh CR. Underrepresentation of Med. 2010;268:40–49. doi: 10.1111/j.1365-2796.2009.02204.x.
renal disease in randomized controlled trials of cardiovascular disease. 16. Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White
JAMA. 2006;296:1377–1384. doi: 10.1001/jama.296.11.1377. HD; Joint ESC/ACCF/AHA/WHF Task Force for the Universal
6. Charytan D, Kuntz RE. The exclusion of patients with chronic kid- Definition of Myocardial Infarction. Third universal definition of myo-
ney disease from clinical trials in coronary artery disease. Kidney Int. cardial infarction. Circulation. 2012;126:2020–2035. doi: 10.1161/
2006;70:2021–2030. doi: 10.1038/sj.ki.5001934. CIR.0b013e31826e1058.
7. National Kidney Foundation. K/DOQI clinical practice guidelines for 17. Wu AHB, Jaffe AS, Apple FS, Jesse RL, Francis GL, Morrow DA, Newby
chronic kidney disease: evaluation, classification, and stratification. Am J LK, Ravkilde J, Tang WHW, Christenson RH, Cannon CP, Storrow AB.
Kidney Dis. 2002;39:S1–S266. National Academy of Clinical Biochemistry laboratory medicine prac-
8. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work tice guidelines: use of cardiac troponin and B-type natriuretic peptide or
Group. KDIGO 2012 clinical practice guideline for the evaluation and N-terminal proB-type natriuretic peptide for etiologies other than acute
management of chronic kidney disease. Kidney Int Suppl. 2013;3:S1–S150. coronary syndromes and heart failure. Clin Chem. 2007;53:2086–2096.
9. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney 18. Newby LK, Jesse RL, Babb JD, Christenson RH, De Fer TM, Diamond
disease and the risks of death, cardiovascular events, and hospitalization. GA, Fesmire FM, Geraci SA, Gersh BJ, Larsen GC, Kaul S, McKay
N Engl J Med. 2004;351:1296–1305. doi: 10.1056/NEJMoa041031. CR, Philippides GJ, Weintraub WS. ACCF 2012 expert consensus docu-
10. Tonelli M, Muntner P, Lloyd A, Manns BJ, Klarenbach S, Pannu N, James ment on practical clinical considerations in the interpretation of troponin
MT, Hemmelgarn BR; Alberta Kidney Disease Network. Risk of coronary elevations: a report of the American College of Cardiology Foundation
events in people with chronic kidney disease compared with those with task force on Clinical Expert Consensus Documents. J Am Coll Cardiol.
diabetes: a population-level cohort study. Lancet. 2012;380:807–814. doi: 2012;60:2427–2463. doi: 10.1016/j.jacc.2012.08.969.
10.1016/S0140-6736(12)60572-8. 19. Berger AK, Duval S, Krumholz HM. Aspirin, beta-blocker, and angio-
11. Herzog CA, Littrell K, Arko C, Frederick PD, Blaney M. Clinical char- tensin-converting enzyme inhibitor therapy in patients with end-stage
acteristics of dialysis patients with acute myocardial infarction in the renal disease and an acute myocardial infarction. J Am Coll Cardiol.
United States: a collaborative project of the United States Renal Data 2003;42:201–208.
System and the National Registry of Myocardial Infarction. Circulation. 20. Beattie JN, Soman SS, Sandberg KR, Yee J, Borzak S, Garg M, McCullough
2007;116:1465–1472. doi: 10.1161/CIRCULATIONAHA.107.696765. PA. Determinants of mortality after myocardial infarction in patients with
1146  Circulation  March 24, 2015

advanced renal dysfunction. Am J Kidney Dis. 2001;37:1191–1200. doi: failure and ST-elevation acute myocardial infarction? Am J Cardiol.
10.1053/ajkd.2001.24522. 2006;97:1142–1145. doi: 10.1016/j.amjcard.2005.11.028.
21. McCullough PA, Sandberg KR, Borzak S, Hudson MP, Garg M, Manley 35. Brass LM, Lichtman JH, Wang Y, Gurwitz JH, Radford MJ, Krumholz
HJ. Benefits of aspirin and beta-blockade after myocardial infarction in HM. Intracranial hemorrhage associated with thrombolytic therapy
patients with chronic kidney disease. Am Heart J. 2002;144:226–232. for elderly patients with acute myocardial infarction: results from the
22. Nyman HA, Dowling TC, Hudson JQ, Peter WL, Joy MS, Nolin TD. Cooperative Cardiovascular Project. Stroke. 2000;31:1802–1811.
Comparative evaluation of the Cockcroft-Gault Equation and the 36. Simoons ML, Maggioni AP, Knatterud G, Leimberger JD, de Jaegere
Modification of Diet in Renal Disease (MDRD) study equation for drug P, van Domburg R, Boersma E, Franzosi MG, Califf R, Schröder R.
dosing: an opinion of the Nephrology Practice and Research Network Individual risk assessment for intracranial haemorrhage during thrombo-
of the American College of Clinical Pharmacy. Pharmacotherapy. lytic therapy. Lancet. 1993;342:1523–1528.
2011;31:1130–1144. doi: 10.1592/phco.31.11.1130. 37. Antithrombotic Trialists’ Collaboration. Collaborative meta-analysis of
23. Melloni C, Peterson ED, Chen AY, Szczech LA, Newby LK, Harrington randomised trials of antiplatelet therapy for prevention of death, myo-
RA, Gibler WB, Ohman EM, Spinler SA, Roe MT, Alexander KP. cardial infarction, and stroke in high risk patients [published correction
Cockcroft-Gault versus modification of diet in renal disease: importance of appears in BMJ. 2002;324:141]. BMJ. 2002;324:71–86.
glomerular filtration rate formula for classification of chronic kidney dis- 38. Yan AT, Yan RT, Tan M, Constance C, Lauzon C, Zaltzman J, Wald R,
ease in patients with non-ST-segment elevation acute coronary syndromes. Fitchett D, Langer A, Goodman SG; Canadian Acute Coronary Syndromes
J Am Coll Cardiol. 2008;51:991–996. doi: 10.1016/j.jacc.2007.11.045. (ACS) Registry Investigators. Treatment and one-year outcome of patients
24. Wargo KA, Eiland EH 3rd, Hamm W, English TM, Phillippe HM.
with renal dysfunction across the broad spectrum of acute coronary syn-
Comparison of the modification of diet in renal disease and Cockcroft- dromes. Can J Cardiol. 2006;22:115–120.
Gault equations for antimicrobial dosage adjustments. Ann Pharmacother. 39. Sciahbasi A, Arcieri R, Quarto M, Pendenza G, Lanzillo C, Summaria F,
2006;40:1248–1253. doi: 10.1345/aph.1G635. Romagnoli E, Commisso C, Penco M, Lioy E. Impact of chronic aspi-
25. Golik MV, Lawrence KR. Comparison of dosing recommendations for rin and statin therapy on presentation of patients with acute myocardial
antimicrobial drugs based on two methods for assessing kidney func- infarction and impaired renal function. Prev Cardiol. 2010;13:18–22. doi:
tion: Cockcroft-Gault and Modification of Diet in Renal Disease. 10.1111/j.1751-7141.2009.00050.x.
Pharmacotherapy. 2008;28:1125–1132. doi: 10.1592/phco.28.9.1125. 40. Baigent C, Landray M, Leaper C, Altmann P, Armitage J, Baxter A, Cairns
26. O’Gara PT, Kushner FG, Ascheim DD, Casey DE Jr, Chung MK, de HS, Collins R, Foley RN, Frighi V, Kourellias K, Ratcliffe PJ, Rogerson M,
Lemos JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, Granger Scoble JE, Tomson CR, Warwick G, Wheeler DC. First United Kingdom
CB, Krumholz HM, Linderbaum JA, Morrow DA, Newby LK, Ornato JP, Heart and Renal Protection (UK-HARP-I) study: biochemical efficacy and
Ou N, Radford MJ, Tamis-Holland JE, Tommaso CL, Tracy CM, Woo safety of simvastatin and safety of low-dose aspirin in chronic kidney dis-
YJ, Zhao DX, Anderson JL, Jacobs AK, Halperin JL, Albert NM, Brindis ease. Am J Kidney Dis. 2005;45:473–484. doi: 10.1053/j.ajkd.2004.11.015.
RG, Creager MA, DeMets D, Guyton RA, Hochman JS, Kovacs RJ, 41. Ethier J, Bragg-Gresham JL, Piera L, Akizawa T, Asano Y, Mason N,
Kushner FG, Ohman EM, Stevenson WG, Yancy CW; American College Gillespie BW, Young EW. Aspirin prescription and outcomes in hemodial-
of Cardiology Foundation/American Heart Association Task Force on ysis patients: the Dialysis Outcomes and Practice Patterns Study (DOPPS).
Practice Guidelines. 2013 ACCF/AHA guideline for the management Am J Kidney Dis. 2007;50:602–611. doi: 10.1053/j.ajkd.2007.07.007.
of ST-elevation myocardial infarction: a report of the American College 42. Jneid H, Anderson JL, Wright RS, Adams CD, Bridges CR, Casey DE
of Cardiology Foundation/American Heart Association Task Force on Jr, Ettinger SM, Fesmire FM, Ganiats TG, Lincoff AM, Peterson ED,
Practice Guidelines. Circulation. 2013;127:e362–e425. doi: 10.1161/ Philippides GJ, Theroux P, Wenger NK, Zidar JP. 2012 ACCF/AHA
Downloaded from http://ahajournals.org by on January 29, 2019

CIR.0b013e3182742cf6. focused update of the Guideline for the Management of Patients With
27. Anderson JL, Adams CD, Antman EM, Bridges CR, Califf RM, Casey Unstable Angina/Non–ST-Elevation Myocardial Infarction (updating the
DE Jr, Chavey WE 2nd, Fesmire FM, Hochman JS, Levin TN, Lincoff 2007 guideline and replacing the 2011 focused update): a report of the
AM, Peterson ED, Theroux P, Wenger NK, Wright RS, Smith SC Jr. 2011 American College of Cardiology Foundation/American Heart Association
ACCF/AHA focused update incorporated into the ACC/AHA 2007 guide- Task Force on Practice Guidelines. Circulation. 2012;126:875–910. doi:
lines for the management of patients with unstable angina/non-ST-eleva- 10.1161/CIR.0b013e318256f1e0.
tion myocardial infarction: a report of the American College of Cardiology 43. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK;

Foundation/American Heart Association Task Force on Practice Guidelines. Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial
Circulation. 2011;123:e426–e579. doi: 10.1161/CIR.0b013e318212bb8b. Investigators. Effects of clopidogrel in addition to aspirin in patients with
28. Angiomax (bivalirudin) injection [package insert]. Parsippany, NJ: The acute coronary syndromes without ST-segment elevation. N Engl J Med.
Medicines Co; 2013. 2001;345:494–502. doi: 10.1056/NEJMoa010746.
29. Melloni C, Roe MT, Chen AY, Wang TY, Wiviott SD, Ho PM, Peterson 44. Keltai M, Tonelli M, Mann JF, Sitkei E, Lewis BS, Hawken S, Mehta
ED, Alexander KP. Use of early clopidogrel by reperfusion strategy SR, Yusuf S; CURE Trial Investigators. Renal function and outcomes in
among patients presenting with ST-segment elevation myocardial infarc- acute coronary syndrome: impact of clopidogrel. Eur J Cardiovasc Prev
tion. Circ Cardiovasc Qual Outcomes. 2011;4:603–609. doi: 10.1161/ Rehabil. 2007;14:312–318. doi: 10.1097/01.hjr.0000220582.19516.a6.
CIRCOUTCOMES.111.961045. 45. Steinhubl SR, Berger PB, Mann JT 3rd, Fry ET, DeLago A, Wilmer C,
30. Gibson CM, Pinto DS, Murphy SA, Morrow DA, Hobbach HP, Wiviott Topol EJ; CREDO Investigators: Clopidogrel for the Reduction of Events
SD, Giugliano RP, Cannon CP, Antman EM, Braunwald E; TIMI Study During Observation. Early and sustained dual oral antiplatelet therapy
Group. Association of creatinine and creatinine clearance on presenta- following percutaneous coronary intervention: a randomized controlled
tion in acute myocardial infarction with subsequent mortality. J Am Coll trial [published correction appears in JAMA. 2003;289:987]. JAMA.
Cardiol. 2003;42:1535–1543. 2002;288:2411–2420.
31. Hobbach HP, Gibson CM, Giugliano RP, Hundertmark J, Schaeffer C, 46. Best PJ, Steinhubl SR, Berger PB, Dasgupta A, Brennan DM, Szczech
Tscherleniak W, Schuster P. The prognostic value of serum creatinine LA, Califf RM, Topol EJ; CREDO Investigators. The efficacy and safety
on admission in fibrinolytic-eligible patients with acute myocardial of short- and long-term dual antiplatelet therapy in patients with mild or
infarction. J Thromb Thrombolysis. 2003;16:167–174. doi: 10.1023/B: moderate chronic kidney disease: results from the Clopidogrel for the
THRO.0000024055.13207.50. Reduction of Events During Observation (CREDO) trial. Am Heart J.
32. Keough-Ryan TM, Kiberd BA, Dipchand CS, Cox JL, Rose CL, Thompson 2008;155:687–693. doi: 10.1016/j.ahj.2007.10.046.
KJ, Clase CM. Outcomes of acute coronary syndrome in a large Canadian 47. Sabatine MS, Cannon CP, Gibson CM, López-Sendón JL, Montalescot
cohort: impact of chronic renal insufficiency, cardiac interventions, and ane- G, Theroux P, Claeys MJ, Cools F, Hill KA, Skene AM, McCabe CH,
mia. Am J Kidney Dis. 2005;46:845–855. doi: 10.1053/j.ajkd.2005.07.043. Braunwald E; CLARITY-TIMI 28 Investigators. Addition of clopido-
33. Medi C, Montalescot G, Budaj A, Fox KA, López-Sendón J, FitzGerald grel to aspirin and fibrinolytic therapy for myocardial infarction with
G, Brieger DB; GRACE Investigators. Reperfusion in patients with renal ST-segment elevation. N Engl J Med. 2005;352:1179–1189. doi: 10.1056/
dysfunction after presentation with ST-segment elevation or left bundle NEJMoa050522.
branch block: GRACE (Global Registry of Acute Coronary Events). JACC 48. Ahmed S, Michael Gibson C, Cannon CP, Murphy SA, Sabatine MS.
Cardiovasc Interv. 2009;2:26–33. doi: 10.1016/j.jcin.2008.09.010. Impact of reduced glomerular filtration rate on outcomes in patients with
34. Dragu R, Behar S, Sandach A, Boyko V, Kapeliovich M, Rispler S, ST-segment elevation myocardial infarction undergoing fibrinolysis: a
Hammerman H. Should primary percutaneous coronary intervention CLARITY-TIMI 28 analysis. J Thromb Thrombolysis. 2011;31:493–500.
be the preferred method of reperfusion therapy for patients with renal doi: 10.1007/s11239-011-0566-9.
Washam et al   Pharmacotherapy in CKD Patients With ACS   1147

49. Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W,


65. Spinler SA, Inverso SM, Cohen M, Goodman SG, Stringer KA, Antman EM;
Gottlieb S, Neumann FJ, Ardissino D, De Servi S, Murphy SA, Riesmeyer J, ESSENCE and TIMI 11B Investigators. Safety and efficacy of unfraction-
Weerakkody G, Gibson CM, Antman EM; TRITON-TIMI 38 Investigators. ated heparin versus enoxaparin in patients who are obese and patients with
Prasugrel versus clopidogrel in patients with acute coronary syndromes. N severe renal impairment: analysis from the ESSENCE and TIMI 11B stud-
Engl J Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482. ies. Am Heart J. 2003;146:33–41. doi: 10.1016/S0002-8703(03)00121-2.
50. Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, 66. Collet JP, Montalescot G, Agnelli G, Van de Werf F, Gurfinkel EP, López-
Horrow J, Husted S, James S, Katus H, Mahaffey KW, Scirica BM, Skene A, Sendón J, Laufenberg CV, Klutman M, Gowda N, Gulba D; GRACE
Steg PG, Storey RF, Harrington RA, Freij A, Thorsén M; PLATO Investigators. Investigators. Non-ST-segment elevation acute coronary syndrome in
Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N patients with renal dysfunction: benefit of low-molecular-weight heparin
Engl J Med. 2009;361:1045–1057. doi: 10.1056/NEJMoa0904327. alone or with glycoprotein IIb/IIIa inhibitors on outcomes: the Global
51. James S, Budaj A, Aylward P, Buck KK, Cannon CP, Cornel JH,
Registry of Acute Coronary Events. Eur Heart J. 2005;26:2285–2293. doi:
Harrington RA, Horrow J, Katus H, Keltai M, Lewis BS, Parikh K, Storey 10.1093/eurheartj/ehi337.
RF, Szummer K, Wojdyla D, Wallentin L. Ticagrelor versus clopidogrel 67. Fox KA, Antman EM, Montalescot G, Agewall S, SomaRaju B, Verheugt
in acute coronary syndromes in relation to renal function: results from FW, Lopez-Sendon J, Hod H, Murphy SA, Braunwald E. The impact of
the Platelet Inhibition and Patient Outcomes (PLATO) trial. Circulation. renal dysfunction on outcomes in the ExTRACT-TIMI 25 trial. J Am Coll
2010;122:1056–1067. doi: 10.1161/CIRCULATIONAHA.109.933796. Cardiol. 2007;49:2249–2255. doi: 10.1016/j.jacc.2006.12.049.
52. Bhatt DL, Fox KA, Hacke W, Berger PB, Black HR, Boden WE, Cacoub 68. Spinler SA, Mahaffey KW, Gallup D, Levine GN, Ferguson JJ 3rd, Rao
P, Cohen EA, Creager MA, Easton JD, Flather MD, Haffner SM, Hamm SV, Gallo R, Ducas J, Goodman SG, Antman E, White HD, Biasucci L,
CW, Hankey GJ, Johnston SC, Mak KH, Mas JL, Montalescot G, Pearson Becker RC, Col JJ, Cohen M, Harrington RA, Califf RM; SYNERGY
TA, Steg PG, Steinhubl SR, Weber MA, Brennan DM, Fabry-Ribaudo L, Trial Investigators. Relationship between renal function and outcomes
Booth J, Topol EJ; CHARISMA Investigators. Clopidogrel and aspirin in high-risk patients with non-ST-segment elevation acute coronary syn-
versus aspirin alone for the prevention of atherothrombotic events. N Engl dromes: results from SYNERGY. Int J Cardiol. 2010;144:36–41. doi:
J Med. 2006;354:1706–1717. doi: 10.1056/NEJMoa060989. 10.1016/j.ijcard.2009.03.119.
53. Dasgupta A, Steinhubl SR, Bhatt DL, Berger PB, Shao M, Mak KH, Fox 69. Fox KA, Bassand JP, Mehta SR, Wallentin L, Theroux P, Piegas LS,
KA, Montalescot G, Weber MA, Haffner SM, Dimas AP, Steg PG, Topol Valentin V, Moccetti T, Chrolavicius S, Afzal R, Yusuf S; OASIS 5
EJ; CHARISMA Investigators. Clinical outcomes of patients with diabetic Investigators. Influence of renal function on the efficacy and safety of
nephropathy randomized to clopidogrel plus aspirin versus aspirin alone fondaparinux relative to enoxaparin in non ST-segment elevation acute
(a post hoc analysis of the clopidogrel for high atherothrombotic risk and coronary syndromes. Ann Intern Med. 2007;147:304–310.
ischemic stabilization, management, and avoidance [CHARISMA] trial). 70. Becker RC, Spencer FA, Gibson M, Rush JE, Sanderink G, Murphy SA,
Am J Cardiol. 2009;103:1359–1363. doi: 10.1016/j.amjcard.2009.01.342. Ball SP, Antman EM; TIMI 11A Investigators. Influence of patient charac-
54. Integrilin (eptifibatide) injection [product information]. Whitehouse teristics and renal function on factor Xa inhibition pharmacokinetics and
Station, NJ: Merck & Co, Inc; 2014. pharmacodynamics after enoxaparin administration in non-ST-segment
55. Aggrastat (tirofiban) injection [product information]. Somerset, NJ: elevation acute coronary syndromes. Am Heart J. 2002;143:753–759.
Medicure Pharma, Inc; 2014. 71. Lim W, Dentali F, Eikelboom JW, Crowther MA. Meta-analysis: low-
56. Januzzi JL Jr, Snapinn SM, DiBattiste PM, Jang IK, Theroux P. Benefits molecular-weight heparin and bleeding in patients with severe renal insuf-
and safety of tirofiban among acute coronary syndrome patients with ficiency. Ann Intern Med. 2006;144:673–684.
mild to moderate renal insufficiency: results from the Platelet Receptor 72. LaPointe NM, Chen AY, Alexander KP, Roe MT, Pollack CV Jr, Lytle BL,
Downloaded from http://ahajournals.org by on January 29, 2019

Inhibition in Ischemic Syndrome Management in Patients Limited Ohman ME, Gibler BW, Peterson ED. Enoxaparin dosing and associated
by Unstable Signs and Symptoms (PRISM-PLUS) trial. Circulation. risk of in-hospital bleeding and death in patients with non ST-segment ele-
2002;105:2361–2366. vation acute coronary syndromes. Arch Intern Med. 2007;167:1539–1544.
57. Best PJ, Lennon R, Gersh BJ, Ting HH, Rihal CS, Bell MR, Herzog doi: 10.1001/archinte.167.14.1539.
CA, Holmes DR Jr, Berger PB. Safety of abciximab in patients with 73. Anderson JL, Adams CD, Antman EM, Bridges CR, Califf RM, Casey
chronic renal insufficiency who are undergoing percutaneous coro- DE Jr, Chavey WE 2nd, Fesmire FM, Hochman JS, Levin TN, Lincoff
nary interventions. Am Heart J. 2003;146:345–350. doi: 10.1016/ AM, Peterson ED, Theroux P, Wenger NK, Wright RS. ACC/AHA 2007
S0002-8703(03)00231-X. guidelines for the management of patients with unstable angina/non–ST-
58. Reddan DN, O’Shea JC, Sarembock IJ, Williams KA, Pieper KS, Santoian elevation myocardial infarction: a report of the American College of
E, Owen WF, Kitt MM, Tcheng JE. Treatment effects of eptifibatide in Cardiology/American Heart Association Task Force on Practice Guidelines
planned coronary stent implantation in patients with chronic kidney dis- (Writing Committee to Revise the 2002 Guidelines for the Management of
ease (ESPRIT Trial). Am J Cardiol. 2003;91:17–21. Patients With Unstable Angina/Non–ST-Elevation Myocardial Infarction).
59. Freeman RV, Mehta RH, Al Badr W, Cooper JV, Kline-Rogers E, Eagle Circulation. 2007;116:e148–304.
KA. Influence of concurrent renal dysfunction on outcomes of patients 74. Samama MM, Gerotziafas GT. Evaluation of the pharmacological proper-
with acute coronary syndromes and implications of the use of glycopro- ties and clinical results of the synthetic pentasaccharide (fondaparinux).
tein IIb/IIIa inhibitors. J Am Coll Cardiol. 2003;41:718–724. Thromb Res. 2003;109:1–11.
60. Frilling B, Zahn R, Fraiture B, Mark B, Dönges K, Becker T, Siegler KE, 75. Fifth Organization to Assess Strategies in Acute Ischemic Syndromes
Seidl K, Rustige J, Senges J. Comparison of efficacy and complication rates Investigators, Yusuf S, Mehta SR, Chrolavicius S, Afzal R, Pogue J,
after percutaneous coronary interventions in patients with and without renal Granger CB, Budaj A, Peters RJG, Bassand J-P, Wallentin L, Joyner C,
insufficiency treated with abciximab. Am J Cardiol. 2002;89:450–452. Fox KAA. Comparison of fondaparinux and enoxaparin in acute coronary
61. Berger PB, Best PJ, Topol EJ, White J, DiBattiste PM, Chan AW, Kristensen syndromes. N Engl J Med. 2006;354:1464–1476.
SD, Herrmann HC, Moliterno DJ. The relation of renal function to ischemic 76. Lui HK. Dosage, pharmacological effects and clinical outcomes for

and bleeding outcomes with 2 different glycoprotein IIb/IIIa inhibitors: the bivalirudin in percutaneous coronary intervention. J Invasive Cardiol.
do Tirofiban and ReoPro Give Similar Efficacy Outcome (TARGET) trial. 2000;(suppl F):41F–52F.
Am Heart J. 2005;149:869–875. doi: 10.1016/j.ahj.2004.12.002. 77. Robson RA. The influence of dose, gender and kidney function on bivali-
62. Melloni C, James SK, White JA, Giugliano RP, Harrington RA, Huber rudin pharmacokinetics and pharmacodynamics in patients undergoing
K, Tricoci P, Armstrong PW, Van de Werf F, Montalescot G, Califf RM, percutaneous transluminal angioplasty [abstract]. Cambridge, MA; The
Newby LK. Safety and efficacy of adjusted-dose eptifibatide in patients Medicines Company; 2000. Data on file.
with acute coronary syndromes and reduced renal function. Am Heart J. 78. Scica D. A pharmacokinetic and pharmacodynamic study of BG8967 in
2011;162:884–892.e1. doi: 10.1016/j.ahj.2011.08.020. subjects with renal insufficiency: Study C93-313. Cambridge, MA; The
63. Oler A, Whooley MA, Oler J, Grady D. Adding heparin to aspirin reduces Medicines Company; 2000. Data on file.
the incidence of myocardial infarction and death in patients with unstable 79. Chew DP, Bhatt DL, Kimball W, Henry TD, Berger P, McCullough PA,
angina: a meta-analysis. JAMA. 1996;276:811–815. Feit F, Bittl JA, Lincoff AM. Bivalirudin provides increasing benefit with
64. Hirsh J, Bauer KA, Donati MB, Gould M, Samama MM, Weitz JI;
decreasing renal function: a meta-analysis of randomized trials. Am J
American College of Chest Physicians. Parenteral anticoagulants: Cardiol. 2003;92:919–923.
American College of Chest Physicians Evidence-Based Clinical Practice 80. Chew DP, Lincoff AM, Gurm H, Wolski K, Cohen DJ, Henry T, Feit F,
Guidelines (8th Edition). Chest. 2008;133(suppl):141S–159S. doi: Topol EJ; REPLACE-2 Investigators. Bivalirudin versus heparin and glyco-
10.1378/chest.08-0689. protein IIb/IIIa inhibition among patients with renal impairment undergoing
1148  Circulation  March 24, 2015

percutaneous coronary intervention (a subanalysis of the REPLACE-2 trial).   95. Bae EH, Lim SY, Cho KH, Choi JS, Kim CS, Park JW, Ma SK, Jeong
Am J Cardiol. 2005;95:581–585. doi: 10.1016/j.amjcard.2004.11.003. MH, Kim SW. GFR and cardiovascular outcomes after acute myocardial
81. Delhaye C, Maluenda G, Wakabayashi K, Ben-Dor I, Collins SD, Syed AI, infarction: results from the Korea Acute Myocardial Infarction Registry.
Gonzalez MA, Gaglia MA, Torguson R, Xue Z, Suddath WO, Satler LF, Kent Am J Kidney Dis. 2012;59:795–802. doi: 10.1053/j.ajkd.2012.01.016.
KM, Lindsay J, Pichard AD, Waksman R. Safety and in-hospital outcomes of  96. ISIS-4 (Fourth International Study of Infarct Survival) Collaborative
bivalirudin use in dialysis patients undergoing percutaneous coronary interven- Group. ISIS-4: a randomised factorial trial assessing early oral captopril,
tion. Am J Cardiol. 2010;105:297–301. doi: 10.1016/j.amjcard.2009.09.030. oral mononitrate, and intravenous magnesium sulphate in 58,050 patients
82. Mehran R, Nikolsky E, Lansky AJ, Kirtane AJ, Kim YH, Feit F, Manoukian with suspected acute myocardial infarction. Lancet. 1995;345:669–685.
S, Moses JW, Ebrahimi R, Ohman EM, White HD, Pocock SJ, Dangas  97. Gruppo Italiano per lo Studio della Sopravvivenza nell’infarto
GD, Stone GW. Impact of chronic kidney disease on early (30-day) and Miocardico. GISSI-3: effects of lisinopril and transdermal glyceryl trini-
late (1-year) outcomes of patients with acute coronary syndromes treated trate singly and together on 6-week mortality and ventricular function
with alternative antithrombotic treatment strategies: an ACUITY (Acute after acute myocardial infarction. Lancet. 1994;343:1115–1122.
Catheterization and Urgent Intervention Triage strategY) substudy. JACC  98. Morrison LJ, Verbeek PR, McDonald AC, Sawadsky BV, Cook DJ.
Cardiovasc Interv. 2009;2:748–757. doi: 10.1016/j.jcin.2009.05.018. Mortality and prehospital thrombolysis for acute myocardial infarction:
83. Saltzman AJ, Stone GW, Claessen BE, Narula A, Leon-Reyes S, Weisz G, a meta-analysis. JAMA. 2000;283:2686–2692.
Brodie B, Witzenbichler B, Guagliumi G, Kornowski R, Dudek D,  99. Dickstein K, Kjekshus J; OPTIMAAL Steering Committee of the
Metzger DC, Lansky AJ, Nikolsky E, Dangas GD, Mehran R. Long- OPTIMAAL Study Group. Effects of losartan and captopril on mortality
term impact of chronic kidney disease in patients with ST-segment and morbidity in high-risk patients after acute myocardial infarction: the
elevation myocardial infarction treated with primary percutaneous coro- OPTIMAAL randomised trial. Optimal Trial in Myocardial Infarction
nary intervention: the HORIZONS-AMI (Harmonizing Outcomes With with Angiotensin II Antagonist Losartan. Lancet. 2002;360:752–760.
Revascularization and Stents in Acute Myocardial Infarction) trial. JACC 100. Pfeffer MA, McMurray JJ, Velazquez EJ, Rouleau JL, Køber L,

Cardiovasc Interv. 2011;4:1011–1019. doi: 10.1016/j.jcin.2011.06.012. Maggioni AP, Solomon SD, Swedberg K, Van de Werf F, White H,
84. Stone GW, Witzenbichler B, Guagliumi G, Peruga JZ, Brodie BR, Dudek Leimberger JD, Henis M, Edwards S, Zelenkofske S, Sellers MA,
D, Kornowski R, Hartmann F, Gersh BJ, Pocock SJ, Dangas G, Wong SC, Califf RM; Valsartan in Acute Myocardial Infarction Trial Investigators.
Kirtane AJ, Parise H, Mehran R; HORIZONS-AMI Trial Investigators. Valsartan, captopril, or both in myocardial infarction complicated
Bivalirudin during primary PCI in acute myocardial infarction. N Engl J by heart failure, left ventricular dysfunction, or both. N Engl J Med.
Med. 2008;358:2218–2230. doi: 10.1056/NEJMoa0708191. 2003;349:1893–1906. doi: 10.1056/NEJMoa032292.
85. Antman EM, Morrow DA, McCabe CH, Murphy SA, Ruda M, Sadowski 101. Krumholz HM, Anderson JL, Bachelder BL, Fesmire FM, Fihn SD,
Z, Budaj A, López-Sendón JL, Guneri S, Jiang F, White HD, Fox KA, Foody JM, Ho PM, Kosiborod MN, Masoudi FA, Nallamothu BK. ACC/
Braunwald E; ExTRACT-TIMI 25 Investigators. Enoxaparin ver- AHA 2008 performance measures for adults with ST-elevation and non-
sus unfractionated heparin with fibrinolysis for ST-elevation myocar- ST-elevation myocardial infarction: a report of the American College
dial infarction. N Engl J Med. 2006;354:1477–1488. doi: 10.1056/ of Cardiology/American Heart Association Task Force on Performance
NEJMoa060898. Measures (Writing Committee to Develop Performance Measures for
86. Wright RS,Anderson JL, Adams CD, Bridges CR, Casey DE Jr, Ettinger ST-Elevation and Non-ST-Elevation Myocardial Infarction). Circulation.
SM, Fesmire FM, Ganiats TG, Jneid H, Lincoff AM, Peterson ED, 2008;118:2596–2648. doi: 10.1161/CIRCULATIONAHA.108.1911099.
Philippides GJ, Theroux P, Wenger NK,Zidar JP. 2011 ACCF/AHA 102. Han JH, Chandra A, Mulgund J, Roe MT, Peterson ED, Szczech LA,
focused update incorporated into the ACC/AHA 2007 guidelines for the Patel U, Ohman EM, Lindsell CJ, Gibler WB. Chronic kidney disease in
Downloaded from http://ahajournals.org by on January 29, 2019

management of patients with unstable angina/non-ST-elevation myocar- patients with non-ST-segment elevation acute coronary syndromes. Am
dial infarction: a report of the American College of Cardiology Foundation/ J Med. 2006;119:248–254. doi: 10.1016/j.amjmed.2005.08.057.
American Heart Association Task Force on Practice Guidelines. J Am Coll 103. Reddan DN, Szczech L, Bhapkar MV, Moliterno DJ, Califf RM, Ohman
Cardiol. 2011;57:e215–e367. doi: 10.1016/j.jacc.2011.02.011. EM, Berger PB, Hochman JS, Van de Werf F, Harrington RA, Newby
87. Antman EM, Hand M, Armstrong PW, Bates ER, Green LA, Halasyamani LK. Renal function, concomitant medication use and outcomes follow-
LK, Hochman JS, Krumholz HM, Lamas GA, Mullany CJ, Pearle DL, ing acute coronary syndromes. Nephrol Dial Transplant. 2005;20:2105–
Sloan MA, Smith SC Jr. 2007 Focused update of the ACC/AHA 2004 2112. doi: 10.1093/ndt/gfh981.
Guidelines for the Management of Patients With ST-Elevation Myocardial 104. Roy P, Bouchard J, Amyot R, Madore F. Prescription patterns of pharma-
Infarction: a report of the American College of Cardiology/American cological agents for left ventricular systolic dysfunction among hemo-
Heart Association Task Force on Practice Guidelines (Writing Group to dialysis patients. Am J Kidney Dis. 2006;48:645–651. doi: 10.1053/j.
Review New Evidence and Update the ACC/AHA 2004 Guidelines for the ajkd.2006.06.006.
Management of Patients With ST-Elevation Myocardial Infarction) [pub- 105. El-Menyar A, Zubaid M, Sulaiman K, Singh R, Al Thani H, Akbar M,
lished correction appears in J Am Coll Cardiol. 2008;51:977]. J Am Coll Bulbanat B, Al-Hamdan R, Almahmmed W, Al Suwaidi J. In-hospital
Cardiol. 2008;51:210–247. doi: 10.1016/j.jacc.2007.10.001. major clinical outcomes in patients with chronic renal insufficiency
88. First International Study of Infarct Survival Collaborative Group.
presenting with acute coronary syndrome: data from a registry of
Randomised trial of intravenous atenolol among 16 027 cases of suspected 8176 patients. Mayo Clin Proc. 2010;85:332–340. doi: 10.4065/
acute myocardial infarction: ISIS-1. Lancet. 1986;2:57–66. mcp.2009.0513.
89. The MIAMI Trial Research Group. Metoprolol in acute myocardial infarc- 106. Winkelmayer WC, Levin R, Setoguchi S. Associations of kidney func-
tion (MIAMI): a randomised placebo-controlled international trial. Eur tion with cardiovascular medication use after myocardial infarction. Clin
Heart J. 1985;6:199–226. J Am Soc Nephrol. 2008;3:1415–1422. doi: 10.2215/CJN.02010408.
90. Chen ZM, Pan HC, Chen YP, Peto R, Collins R, Jiang LX, Xie JX, Liu 107. Winkelmayer WC, Charytan DM, Brookhart MA, Levin R, Solomon
LS; COMMIT (ClOpidogrel and Metoprolol in Myocardial Infarction DH, Avorn J. Kidney function and use of recommended medications
Trial) Collaborative Group. Early intravenous then oral metopro- after myocardial infarction in elderly patients. Clin J Am Soc Nephrol.
lol in 45,852 patients with acute myocardial infarction: randomised 2006;1:796–801. doi: 10.2215/CJN.00150106.
placebo-controlled trial. Lancet. 2005;366:1622–1632. doi: 10.1016/ 108. Ahmed A, Centor RM, Weaver MT, Perry GJ. A propensity score

S0140-6736(05)67661-1. analysis of the impact of angiotensin-converting enzyme inhibitors
91. Propranolol in acute myocardial infarction: a multicentre trial. Lancet. on long-term survival of older adults with heart failure and perceived
1966;2:1435–1438. contraindications. Am Heart J. 2005;149:737–743. doi: 10.1016/j.
92. Dargie HJ. Effect of carvedilol on outcome after myocardial infarction in ahj.2004.06.030.
patients with left-ventricular dysfunction: the CAPRICORN randomised 109. Ponikowski P, Ronco C, Anker SD. Cardiorenal syndromes: recom-
trial. Lancet. 2001;357:1385–1390. mendations from clinical practice guidelines: the cardiologist’s view.
93. Tenormin oral tablets, atenolol oral tablets [product information].
Contrib Nephrol. 2010;165:145–152. doi: 10.1159/000313753.
Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2012. 110. Ronco C, McCullough P, Anker SD, Anand I, Aspromonte N, Bagshaw
94. Wali RK, Iyengar M, Beck GJ, Chartyan DM, Chonchol M, Lukas MA, SM, Bellomo R, Berl T, Bobek I, Cruz DN, Daliento L, Davenport A,
Cooper C, Himmelfarb J, Weir MR, Berl T, Henrich WL, Cheung AK. Haapio M, Hillege H, House AA, Katz N, Maisel A, Mankad S, Zanco
Efficacy and safety of carvedilol in treatment of heart failure with chronic P, Mebazaa A, Palazzuoli A, Ronco F, Shaw A, Sheinfeld G, Soni S,
kidney disease: a meta-analysis of randomized trials. Circ Heart Fail. Vescovo G, Zamperetti N, Ponikowski P; Acute Dialysis Quality
2011;4:18–26. doi: 10.1161/CIRCHEARTFAILURE.109.932558. Initiative (ADQI) Consensus Group. Cardio-renal syndromes: report
Washam et al   Pharmacotherapy in CKD Patients With ACS   1149

from the Consensus Conference of the Acute Dialysis Quality Initiative. lowering with statins after acute coronary syndromes. N Engl J Med.
Eur Heart J. 2010;31:703–711. doi: 10.1093/eurheartj/ehp507. 2004;350:1495–1504. doi: 10.1056/NEJMoa040583.
111. Bakris GL, Weir MR. Angiotensin-converting enzyme inhibitor-­
127. de Lemos JA, Blazing MA, Wiviott SD, Lewis EF, Fox KA, White
associated elevations in serum creatinine: is this a cause for concern? HD, Rouleau JL, Pedersen TR, Gardner LH, Mukherjee R, Ramsey
Arch Intern Med. 2000;160:685–693. KE, Palmisano J, Bilheimer DW, Pfeffer MA, Califf RM, Braunwald
112. Ronco C, Haapio M, House AA, Anavekar N, Bellomo R. Cardiorenal E; for the A to Z Investigators. Early intensive vs a delayed conserva-
syndrome. J Am Coll Cardiol. 2008;52:1527–1539. doi: 10.1016/j. tive simvastatin strategy in patients with acute coronary syndromes:
jacc.2008.07.051. phase Z of the A to Z trial. JAMA. 2004;292:1307–1316. doi: 10.1001/
113. Garthwaite E, Bhandari S. The effects of angiotensin converting enzyme jama.292.11.1307.
inhibitors on potassium homeostasis in dialysis patients with and 128. Baigent C, Landray MJ, Reith C, Emberson J, Wheeler DC, Tomson
without residual renal function. Artif Organs. 2009;33:641–647. doi: C, Wanner C, Krane V, Cass A, Craig J, Neal B, Jiang L, Hooi LS,
10.1111/j.1525-1594.2009.00790.x. Levin A, Agodoa L, Gaziano M, Kasiske B, Walker R, Massy ZA,
114. Knoll GA, Sahgal A, Nair RC, Graham J, van Walraven C, Burns KD. Feldt-Rasmussen B, Krairittichai U, Ophascharoensuk V, Fellström B,
Renin-angiotensin system blockade and the risk of hyperkalemia in Holdaas H, Tesar V, Wiecek A, Grobbee D, de Zeeuw D, Grönhagen-
chronic hemodialysis patients. Am J Med. 2002;112:110–114. Riska C, Dasgupta T, Lewis D, Herrington W, Mafham M, Majoni W,
115. Han SW, Won YW, Yi JH, Kim HJ. No impact of hyperkalaemia with Wallendszus K, Grimm R, Pedersen T, Tobert J, Armitage J, Baxter A,
renin-angiotensin system blockades in maintenance haemodialysis Bray C, Chen Y, Chen Z, Hill M, Knott C, Parish S, Simpson D, Sleight
patients. Nephrol Dial Transplant. 2007;22:1150–1155. doi: 10.1093/ P, Young A, Collins R; SHARP Investigators. The effects of lowering
ndt/gfl752. LDL cholesterol with simvastatin plus ezetimibe in patients with chronic
116. Zannad F, Kessler M, Lehert P, Grünfeld JP, Thuilliez C, Leizorovicz kidney disease (Study of Heart and Renal Protection): a randomised
A, Lechat P. Prevention of cardiovascular events in end-stage renal placebo-controlled trial. Lancet. 2011;377:2181–2192. doi: 10.1016/
disease: results of a randomized trial of fosinopril and implications S0140-6736(11)60739-3.
for future studies. Kidney Int. 2006;70:1318–1324. doi: 10.1038/ 129. Fellström BC, Jardine AG, Schmieder RE, Holdaas H, Bannister K,
sj.ki.5001657. Beutler J, Chae DW, Chevaile A, Cobbe SM, Grönhagen-Riska C, De
117. Berger WG, Erly WK, Krupinski EA, Standen JR, Stern RG. The soli- Lima JJ, Lins R, Mayer G, McMahon AW, Parving HH, Remuzzi G,
tary pulmonary nodule on chest radiography: can we really tell if the Samuelsson O, Sonkodi S, Sci D, Süleymanlar G, Tsakiris D, Tesar V,
nodule is calcified? AJR Am J Roentgenol. 2001;176:201–204. doi: Todorov V, Wiecek A, Wüthrich RP, Gottlow M, Johnsson E, Zannad
10.2214/ajr.176.1.1760201. F; AURORA Study Group. Rosuvastatin and cardiovascular events in
118. Frances CD, Noguchi H, Massie BM, Browner WS, McClellan M. Are patients undergoing hemodialysis. N Engl J Med. 2009;360:1395–1407.
we inhibited? Renal insufficiency should not preclude the use of ACE doi: 10.1056/NEJMoa0810177.
inhibitors for patients with myocardial infarction and depressed left ven- 130. Holdaas H, Fellström B, Jardine AG, Holme I, Nyberg G, Fauchald P,
tricular function. Arch Intern Med. 2000;160:2645–2650. Grönhagen-Riska C, Madsen S, Neumayer HH, Cole E, Maes B, Ambühl
119. Hillege HL, van Gilst WH, van Veldhuisen DJ, Navis G, Grobbee DE, de P, Olsson AG, Hartmann A, Solbu DO, Pedersen TR; Assessment of
Graeff PA, de Zeeuw D; CATS Randomized Trial. Accelerated decline LEscol in Renal Transplantation (ALERT) Study Investigators. Effect
and prognostic impact of renal function after myocardial infarction and of fluvastatin on cardiac outcomes in renal transplant recipients: a mul-
the benefits of ACE inhibition: the CATS randomized trial. Eur Heart J. ticentre, randomised, placebo-controlled trial. Lancet. 2003;361:2024–
2003;24:412–420. 2031. doi: 10.1016/S0140-6736(03)13638-0.
Downloaded from http://ahajournals.org by on January 29, 2019

120. Krause MW, Massing M, Kshirsagar A, Rosamond W, Simpson RJ Jr. 131. Wanner C, Krane V, März W, Olschewski M, Mann JF, Ruf G, Ritz
Combination therapy improves survival after acute myocardial infarction E; German Diabetes and Dialysis Study Investigators. Atorvastatin in
in the elderly with chronic kidney disease. Ren Fail. 2004;26:715–725. patients with type 2 diabetes mellitus undergoing hemodialysis. N Engl
121. Tokmakova MP, Skali H, Kenchaiah S, Braunwald E, Rouleau JL,
J Med. 2005;353:238–248. doi: 10.1056/NEJMoa043545.
Packer M, Chertow GM, Moyé LA, Pfeffer MA, Solomon SD. Chronic 132. Lim SY, Bae EH, Choi JS, Kim CS, Park JW, Ma SK, Jeong MH, Kim
kidney disease, cardiovascular risk, and response to angiotensin-con- SW; Korea Acute Myocardial Infarction Registry Investigators. Effect
verting enzyme inhibition after myocardial infarction: the Survival And on short- and long-term major adverse cardiac events of statin treatment
Ventricular Enlargement (SAVE) study. Circulation. 2004;110:3667– in patients with acute myocardial infarction and renal dysfunction. Am J
3673. doi: 10.1161/01.CIR.0000149806.01354.BF. Cardiol. 2012;109:1425–1430. doi: 10.1016/j.amjcard.2012.01.351.
122. Shlipak MG, Browner WS, Noguchi H, Massie B, Frances CD,
133. Szummer K, Lundman P, Jacobson SH, Schön S, Lindbäck J, Stenestrand
McClellan M. Comparison of the effects of angiotensin converting- U, Wallentin L, Jernberg T. Association between statin treatment and
enzyme inhibitors and beta blockers on survival in elderly patients with outcome in relation to renal function in survivors of myocardial infarc-
reduced left ventricular function after myocardial infarction. Am J Med. tion. Kidney Int. 2011;79:997–1004. doi: 10.1038/ki.2010.524.
2001;110:425–433. 134. Shibui T, Nakagomi A, Kusama Y, Atarashi H, Mizuno K. Impact of
123. Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Roniker B, Bittman statin therapy on renal function and long-term prognosis in acute
R, Hurley S, Kleiman J, Gatlin M; Eplerenone Post-Acute Myocardial coronary syndrome patients with chronic kidney disease. Int Heart J.
Infarction Heart Failure Efficacy and Survival Study Investigators. 2010;51:312–318.
Eplerenone, a selective aldosterone blocker, in patients with left 135. Tonelli M, Moyé L, Sacks FM, Kiberd B, Curhan G; Cholesterol and
ventricular dysfunction after myocardial infarction. N Engl J Med. Recurrent Events (CARE) Trial Investigators. Pravastatin for secondary
2003;348:1309–1321. doi: 10.1056/NEJMoa030207. prevention of cardiovascular events in persons with mild chronic renal
124. Juurlink DN, Mamdani MM, Lee DS, Kopp A, Austin PC, Laupacis insufficiency. Ann Intern Med. 2003;138:98–104.
A, Redelmeier DA. Rates of hyperkalemia after publication of the 136. McClure DL, Valuck RJ, Glanz M, Murphy JR, Hokanson JE. Statin and
Randomized Aldactone Evaluation Study. N Engl J Med. 2004;351:543– statin-fibrate use was significantly associated with increased myositis
551. doi: 10.1056/NEJMoa040135. risk in a managed care population. J Clin Epidemiol. 2007;60:812–818.
125. Rossignol P, Cleland JG, Bhandari S, Tala S, Gustafsson F, Fay R, doi: 10.1016/j.jclinepi.2006.11.006.
Lamiral Z, Dobre D, Pitt B, Zannad F. Determinants and conse- 137. US Department of Health and Human Services, Food and Drug

quences of renal function variations with aldosterone blocker therapy Administration, Center for Drug Evaluation and Research (CDER).
in heart failure patients after myocardial infarction: insights from the Guidance for industry: pharmacokinetics in patients with impaired renal
Eplerenone Post-Acute Myocardial Infarction Heart Failure Efficacy function: study design, data analysis, and impact on dosing and label-
and Survival Study. Circulation. 2012;125:271–279. doi: 10.1161/ ing. Silver Spring, MD: US Department of Health and Human Services;
CIRCULATIONAHA.111.028282. March 2010. http://www.fda.gov/downloads/Drugs/Guidances/
126. Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL,
UCM204959.pdf. Accessed December 5, 2014.
Belder R, Joyal SV, Hill KA, Pfeffer MA, Skene AM; Pravastatin
or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Key Words: AHA Scientific Statements ◼ acute coronary syndrome ◼
Myocardial Infarction 22 Investigators. Intensive versus moderate lipid chronic kidney disease ◼ kidney disease ◼ pharmacotherapy

You might also like