You are on page 1of 9

In Practice

The Controversy of Contrast-Induced


Nephropathy With Intravenous Contrast:
What Is the Risk?
Michael R. Rudnick, Amanda K. Leonberg-Yoo, Harold I. Litt, Raphael M. Cohen,
Susan Hilton, and Peter P. Reese

Contrast-induced nephropathy (CIN) has long been observed in both experimental and clinical studies. Complete author and article
However, recent observational studies have questioned the prevalence and severity of CIN following information provided before
references.
intravenous contrast exposure. Initial studies of acute kidney injury following intravenous contrast were
limited by the absence of control groups or contained control groups that did not adjust for additional Am J Kidney Dis. 75(1):
acute kidney injury risk factors, including prevalent chronic kidney disease, as well as accepted pro- 105-113. Published online
August 28, 2019.
phylactic strategies. More contemporary use of propensity score–adjusted models have attempted to
minimize the risk for selection bias, although bias cannot be completely eliminated without a pro- doi: 10.1053/
spective randomized trial. Based on existing data, we recommend the following CIN risk classification: j.ajkd.2019.05.022
patients with estimated glomerular filtration rates (eGFRs) ≥ 45 mL/min/1.73 m2 are at negligible risk © 2019 by the National
for CIN, while patients with eGFRs < 30 mL/min/1.73 m2 are at high risk for CIN. Patients with eGFRs Kidney Foundation, Inc.
between 30 and 44 mL/min/1.73 m2 are at an intermediate risk for CIN unless diabetes mellitus is
present, which would further increase the risk. In all patients at any increased risk for CIN, the risk for
CIN needs to be balanced by the risk of not performing an intravenous contrast-enhanced study.

Note from Editors: This article was commis-


glomerular filtration rate (eGFR) was FEATURE EDITOR:
sioned to celebrate the selection of Iodinated 39 mL/min/1.73 m2 (using the CKD-EPI Holly Kramer
Contrast as a finalist in NephMadness 2018. equation). A urinalysis dipstick demon-
NephMadness is an educational project styled strated protein (1+) but no other abnor- ADVISORY BOARD:
as a tournament in which key concepts in malities. A chest x-ray was normal and Linda Fried
nephrology “compete” to determine which de- lower-extremity duplex ultrasonography Ana Ricardo
serves to be crowned the most notable recent Roger Rodby
was negative. Pulmonary embolism was Robert Toto
advance in the field. suspected and a discussion occurred about
the risk for contrast-induced nephropathy In Practice is a
(CIN) with computed tomography angiog- focused review
raphy (CTA). providing in-depth
Clinical Vignette guidance on a clinical
A 60-year-old man presented to the emer- topic that nephrolo-
gency department with sudden onset of Introduction gists commonly
encounter. Using
right anterior chest pain. The pain was sharp Contrast media (CM) was first reported to clinical vignettes,
in nature, exacerbated with breathing, and cause acute kidney injury (AKI) in 1954 when these articles illus-
there were no symptoms of fever, hemop- Bartels et al1 reported a patient with multiple trate a complex prob-
tysis, or shortness of breath. The patient also myeloma who developed anuria following lem for which optimal
provided a history of right calf pain for the intravenous pyelography. This report was diagnostic and/or
therapeutic ap-
past 3 days. Medical history includes hy- followed by additional case series of AKI after proaches are
pertension and diabetes mellitus (DM) each intravascular CM with excretory urography, uncertain.
of 10 years’ duration, and chronic kidney computed tomography (CT), and non-
disease (CKD; baseline serum creatinine coronary angiography.2 In the 1980s, larger
[Scr], 1.9 mg/dL). Physical examination series of AKI following CM administration
revealed a mildly obese man with blood during coronary angiography began to appear,
pressure of 160/100 mm Hg, pulse rate of with CKD and DM being established as risk
95 beats/min, regular respirations with rate factors for this complication.3 The entity of
of 16 breaths/min, temperature of 98.8 F, CIN was coined, most commonly in associa-
and pulse oximetry while breathing room tion with coronary angiography despite 90%
air of 91% oxygen saturation. The rest of of all intravascular CM being given intrave-
the examination findings were normal nously during CT. Until recently it has been
with the exception of a tender nonswollen accepted that in high-risk patients, CM given
right calf. Laboratory results were normal either intra-arterially or intravenously was
except for Scr level of 1.8 mg/dL. Estimated capable of causing AKI. In addition, there is

AJKD Vol 75 | Iss 1 | January 2020 105


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

extensive experimental literature demonstrating contrast Comparison of CIN With Intra-arterial and
nephrotoxicity, supporting the possibility that CM is Intravenous Contrast Administration
nephrotoxic in humans.4-8
Those who propose that CIN from intravenous CM is
During the past 10 to 15 years, an increasing number of
overstated argue that this conclusion has been inappro-
publications have called into question whether CIN
priately extrapolated from studies investigating AKI
following intravenous CM administration has been over-
following intra-arterial CM exposure.9 Given differences in
stated, exists at all, and is clinically relevant.2,9-11 These
baseline patient comorbid conditions and the procedural
opinions arose from controlled studies demonstrating
risks of coronary angiography, it is intuitive to expect a
similar AKI rates in patients who underwent contrast-
higher AKI rate following CM exposure with coronary
enhanced CT (CECT) compared with those who under-
angiography compared to CECT.21,22 Numerous studies of
went unenhanced CT. It is important to determine the true
patients undergoing coronary angiography have demon-
risk for nephrotoxicity following intravenous CM expo-
strated AKI rates of w13%, which exceeds the average AKI
sure. If the risk for CIN is overstated, CM exposure may be
rates of 5% to 6% associated with intravenous CM expo-
needlessly avoided in patients who could clinically benefit
sure.13,23-25
from the improved diagnostic accuracy of CECT.12 How-
One conclusion of these results, although controver-
ever, if the risk for CIN is underestimated, patients may be
sial, is that intra-arterial delivery of CM is more neph-
unnecessarily exposed to a nephrotoxic agent capable of
rotoxic compared to intravenous administration.26-28
causing AKI that may have both short- and long-term
Statements that intra-arterial CM administration presents
clinical implications.
a higher concentration of CM to the kidney are only true
The purpose of this review is to critically examine
when intra-arterial contrast is given in the aorta above
publications about AKI following intravenous CM expo-
or at the level of the renal arteries and possibly with
sure in an effort to better define the true incidence of AKI
high-dose left ventriculography (first-pass renal expo-
due to CM administered intravenously and its clinical
sure).29,30 With infrarenal aortic or coronary artery
relevance.
administration, CM will drain into the venous system
before being delivered to the kidney (second-pass renal
Randomized Studies Demonstrating Differences exposure).30
in AKI After Contrast Exposure It has also been suggested that CM volume administered
Support for the position that intravenous CM can be during coronary angiography is higher compared with the
nephrotoxic comes from prospective randomized intravenous CM volume used for CECT, thus accounting
controlled trials (RCTs) demonstrating different AKI rates for the higher rate of AKI with intra-arterial administra-
between groups who received different CM or who were tion. When contrast dose is expressed as the ratio of grams
or were not exposed to a prophylactic intervention. Risk of iodine to GFR (g-I/GFR) to normalize for the exposure
factors for AKI other than type of CM or prophylactic of CM seen by the kidneys, higher ratios correlate with
intervention should be balanced in RCTs, so differences in nephrotoxicity.25 During coronary angiography, mean g-
AKI incidence should be due to contrast nephrotoxicity. I/GFR dose in patients without and with AKI are 0.7 to 1.0
RCTs have demonstrated that high-osmolal CM have a and 1.0 to 1.8, respectively. CECT studies demonstrate a
greater risk for AKI than low-osmolal CM (LOCM), indi- mean g-I/GFR dose of 0.9.25 Thus, at similar high doses,
cating that high-osmolal CM are nephrotoxic.13-15 How- the risk for AKI following intravenous and intra-arterial
ever, this does not prove that LOCM are nephrotoxic administration may be the same.
because it is possible that the AKI observed in patients Studies comparing AKI following intra-arterial and
exposed to LOCM is due to factors other than CM.14,15 For intravenous exposure in the same patients eliminate patient
a study of a prophylactic intervention to provide support differences in risk factors for AKI because each patient
that CM are nephrotoxic, the intervention would need to serves as his or her own internal control. This approach
show a consistent benefit, with no effect itself on the should capture nephrotoxic differences between intra-
pathogenesis of AKI. arterial and intravenous contrast exposure. In the few
Volume expansion is a prophylactic intervention that studies that have examined patients exposed to both intra-
could meet these criteria.16 Several RCTs comparing arterial and intravenous CM, the rates of AKI were
intravenous fluids with no fluids, normal saline with similar.31-33 However, differences in contrast volume be-
half-normal saline solution, and different rates of saline tween intra-arterial and intravenous CM studies are a
solution infusion have shown reductions in AKI limitation of this conclusion. In summary, reported dif-
post–contrast exposure.17-20 In these studies, the differ- ferences in contrast nephrotoxicity between intravenous
ence in AKI between the intervention and control groups and intra-arterial contrast exposure do not appear to be
could be due to contrast nephrotoxicity, although the due to a greater inherent nephrotoxicity with intra-arterial
ability of intravenous fluids to cause hemodilution of Scr CM exposure but rather are likely due to differences in
level and correct hypotension makes this interpretation population comorbid conditions, procedural risks, and
less certain. contrast volumes.

106 AJKD Vol 75 | Iss 1 | January 2020


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

CECT Studies of CIN Without Control Groups study strongly suggested that AKI observed following CM
exposure cannot automatically be assumed to be due to CIN.
Initial studies of AKI following intravenous CM were
Following these 2 studies, multiple observational
conducted, with rare exception, without control groups
controlled studies of AKI following intravenous CM
(patients who underwent similar radiologic procedures
exposure were performed. For the most part, these studies
without CM administration).34-37 These reports identified
demonstrated an equivalent rate of AKI between CECT and
CKD as an independent risk factor for CIN, and that DM
unenhanced CT groups, leading the authors to conclude
was an additional risk factor when coupled with CKD.
that AKI after intravenous CM exposure is overstated or
Many of these studies focused on hospitalized patients due
nonexistent (Table S1). A recent systematic review and
to the necessity of obtaining 24- to 72-hour postscan Scr
meta-analysis of 28 observational controlled studies
data. The focus on hospitalized patients had the unin-
demonstrated that CECT compared with unenhanced CT
tended consequence of creating ascertainment bias toward
was not significantly associated with AKI (odds ratio [OR],
a sicker population with multiple comorbid conditions
0.94; 95% confidence interval [CI], 0.83-1.07).42
that may have been responsible for the observed AKI risk,
Wilhelm-Leen et al43 performed an analysis of more than
independent of CM exposure.35
5 million inpatient admissions to estimate AKI in patients
Among uncontrolled studies of AKI following intrave-
exposed to CM compared with patients with similar
nous CM exposure, the AKI rate was 5.1%, with a wide
baseline comorbid conditions and severity of illness but
range (0%-21%) across individual studies.23,34,35,38 In
without exposure to CM.43 When adjusted for comorbid
patients with moderate to severe CKD, the AKI rate
conditions, the risk for AKI with CM exposure was 7.4%
following intravenous CM exposure varied from 0% to
lower than with no CM exposure. Because CM is not
42%, with most having rates >5%.23 The wide range of
nephroprotective, these findings indicate the presence of
AKI following intravenous CM exposure in these uncon-
selection bias; patients perceived to be at highest risk for
trolled studies is due to differences in clinical settings, the
CIN may have selectively not been exposed to CECT. All
prevalence of noncontrast AKI risk factors, number of
these controlled studies have major limitations, including a
high-risk patients, definitions of AKI, timing of post-
higher prevalence of CKD in the control patients, limited
contrast Scr determinations, presence of prophylactic
numbers of high-risk patients, and lack of adjustment for
strategies, and type and dose of CM used.23,35,36,38 It is
confounding variables.44-47
difficult to determine the true rate of AKI following
intravenous CM exposure using these uncontrolled studies.
CECT Controlled Studies of CIN With Propensity
CECT Studies of CIN With Nonrandomized Score Adjustments
Control Groups Given the imbalance in confounding covariates and se-
The importance of controls in CM exposure studies was lection bias between CM and control populations, the ideal
largely ignored until the publication of 2 landmark approach to determine CIN following intravenous contrast
observational studies.39,40 Newhouse et al39 performed a would be an RCT in patients undergoing CT with or
retrospective analysis of 32,161 hospitalized patients with without CM enhancement, which would ensure balanced
serial Scr values on 5 consecutive days and with no CM comorbid conditions between groups and nonbiased
exposure to determine the frequency of Scr level changes randomization. For ethical and logistical reasons, it is
over time.39 During the 5-day period, a change in Scr level unlikely that such RCTs will be performed. Recognizing
of at least 25% or 0.4 mg/dL was observed in a significant these limitations, several investigators performed studies
proportion of patients with both normal and abnormal using propensity score matching methods, summarized in
baseline Scr values. This is relevant because CIN is usually Table 1.
defined as an increase in Scr level ≥ 0.5 mg/dL or a 25% Davenport et al48 performed a retrospective study with
increase from baseline value, assessed at 48 hours after 1:1 propensity score matching of AKI risk in patients who
contrast exposure.41 The authors concluded that previous had undergone CECT or unenhanced CT. AKI was defined
studies of intravenous CIN using change in Scr level as the using either AKI Network (Scr ≥ 0.3 mg/dL or 50%) or
sole criterion for defining CIN should be re-evaluated traditional (Scr ≥ 0.5 mg/dL or 25%) criteria. In patients
because attributing these changes as AKI due to CM with baseline Scr levels < 1.5 mg/dL (w90% of all pa-
exposure is a post hoc, ergo propter hoc (after this, tients), AKI rates for the CECT and unenhanced CT groups
therefore because of this) logical fallacy. Thus, an increase were similar (Table 1). In patients with baseline Scr lev-
in Scr level following CM exposure does not prove the Scr els ≥ 1.5 mg/dL, the incidence of AKI in the CECT group
level increase was due to CIN.35 was higher than in the unenhanced CT groups. Davenport
Bruce et al40 performed a large retrospective study in et al49 also examined AKI risk with CECT using stratified
11,588 patients comparing AKI incidence in patients who eGFR as a more accurate reflection of baseline kidney
underwent CECT and unenhanced CT and found that the function, with similar results observed. The majority
incidence of AKI in contrast-exposed patients was similar to (79%) had baseline eGFR ≥ 60 mL/min/1.73 m2, while
nonexposed patients. The inclusion of a control arm in this only 0.66% had a baseline eGFR < 30 mL/min/1.73 m2.

AJKD Vol 75 | Iss 1 | January 2020 107


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

Table 1. Incidence of CIN and Adverse Outcomes Following Intravenous Contrast Exposure in Studies With Propensity Score
Matching for AKI Risk

No. Propensity AKI No. (%) with AKI Distribution by AKI Ratea
Author Analyzed Definition +C −C Kidney Function +C −C
Davenport48 10,121 (+C); T; A T: 1,221 (12.1%) T: 1,259 (12.4%) Scr < 1.5: 94.3% T: 11.8% T: 12.6%
(2013)b 10,121 (−C) A: 835 (8.3%) A: 867 (8.6%) A: 7.1% A: 7.3%
Scr ≥ 1.5: 18.4% T: 14.9%c T: 11.3%
A: 22% A: 19.2%
Scr ≥ 1.6: 13.7% T: 16.9%c T: 11.7%
A: 26.3%c A: 19.7%
Scr ≥ 2.0: 4.8% T: 31.2%c T: 14.5%
A: 43.3%c A: 22.5%
Davenport49 8,826 (+C); A 619 (7.0%) 606 (6.9%) eGFR ≥ 60: 79% 5.4% 5.5%
(2013)b 8,826 (−C) eGFR 45-59: 14% 10.5% 10.8%
eGFR 30-44: 6.2% 16.7%d 14.2%
eGFR < 30: 0.66% 36.4%c 19.4%
McDonald50 10,686 (+C); T 515 (4.8%) 544 (5.1%) Scr < 1.5: 68% 3.0% 3.0%
(2013) 10,686 (−C) Scr 1.5-2.0: 23% 9.0% 9.0%
Scr > 2.0: 9% 10.0% 11.0%
McDonald51 6,254 (+C); T 313 (5%) 325 (5.2%) eGFR ≥ 90: 13.1% 1.2% 1.3%
(2014)e 6,254 (−C) eGFR 60-89: 30.9% 2.1% 2.0%
eGFR 30-59: 44.1% 5.8% 6.2%
eGFR < 30: 11.9% 14% 14%
McDonald52 1,639 (+C); T; A T: 123 (7.5%) T: 132 (8.1%) eGFR 30-59: 74.4% T: 5.0% T: 5.8%
(2015)e 1,639 (−C) A: 215 (13.1%) A: 266 (16.2%) A: 10.0% A: 15.0%
eGFR < 30: 25.6% T: 15.0% T: 15.0%
A: 21.0% A: 20.0%
McDonald53 1,402f (+C); T; A T: 87 (6.2%) T: 131 (8.6%) eGFR ≥ 60: 34% T: 3.8% T: 4.6%
(2017)e 1,524 (−C) A: 179 (12.8%) A: 237 (15.6%) A: 7.8% A: 10.0%
eGFR 30-59: 60.7% T: 7.2% T: 7.9%
A: 15.0% A: 18.0%
eGFR < 30: 9.4% T: 11.0% T: 21.0%
A: 17.0% A: 25.0%
McDonald54 1,508 (+C); T; A T: 254 (16.8%) T: 240 (15.9%) eGFR > 45: 81.1% T: 14.0% T: 14.0%
(2017)e 1,508 (−C) A: 526 (34.9%) A: 546 (34.9%) A: 31.0% A: 34.0%
eGFR ≤ 45: 18.9% T: 29.0% T: 25.0%
A: 50.0% A: 45.0%
Hinson55 7,201 (+C); T; A T: 766 (10.6%) T: 786 (10.9%) eGFR ≥ 60: 87.5% T: 10.9% T: 10.5%
(2017)g 7,201 (−C) A: 488 (6.8%) A: 466 (63.5%) A: 6.1% A: 5.7%
eGFR 45-59: 7.9% T: 10.3% T: 8.0%
A: 12.3% A: 9.2%
eGFR 30-44: 3.3% T: 5.0% T: 9.5%
A: 9.1% A: 12.0%
eGFR < 30: 1.1% T: 7.3% T: 5.1%
A: 10.9% A: 14.1%
Abbreviations and definitions: A, Acute Kidney Injury Network definition of AKI (increase from baseline Scr ≥ 0.3 mg/dL or 50% increase in Scr); AKI, acute kidney
injury; +C, contrast-enhanced computed tomography; −C, unenhanced computed tomography; CECT, contrast-enhanced computed tomography; CIN, contrast-induced
nephropathy; CKD, chronic kidney disease; CT, computed tomography; eGFR, estimated glomerular filtration rate (in mL/min/1.73 m2); Scr, serum creatinine (in mg/dL); T,
traditional definition of AKI (increase from baseline Scr ≥ 0.5 mg/dL or 25%).
a
Stratified: percent cohort.
b
Similar study cohort used in both studies.
c
P < 0.05 between the CECT and unenhanced CT groups.
d
Odds ratio, 1.4 (95% confidence interval, 0.997-1.97).
e
Overlap between original McDonald et al 2013 cohort.50
f
Propensity score matching was done 1:1 for all CKD stages except in patients with CKD stages 4 to 5 who underwent a 1:3 (iodixanol:no contrast) matching.
g
Propensity data for −C provided by author.

The rates of AKI between CECT and unenhanced CT pa- McDonald et al50 also performed a retrospective study
tients were similar in the groups with eGFR ≥ 60 mL/min/ with 1:1 propensity score matching of AKI risk among
1.73 m2 but were significantly higher in patients with patients who had undergone CECT or unenhanced CT. The
eGFR < 30 mL/min/1.73 m2 exposed to CECT (Table 1). risk for AKI for the CECT and unenhanced CT patients were
Among patients with eGFRs of 30 to 44 mL/min/1.73 m2, similar for all cohorts when risk was stratified by baseline
there were nominally greater odds of AKI following CECT Scr level: 3% in each group for low risk (<1.5 mg/dL); 9%
exposure, compared to unenhanced CT imaging (OR, in each group for medium risk (1.5-2.0 mg/dL); and
1.40; 95% CI, 1.00-1.97), although the difference was not 10% versus 11%, respectively, for high risk (>2.0 mg/dL).
statistically significant. In a smaller subset of patients who underwent both

108 AJKD Vol 75 | Iss 1 | January 2020


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

unenhanced CT and CECT, the risk for AKI for the CECT Examining data from RCTs evaluating prophylactic in-
and unenhanced CT groups were also similar: 4.5% and terventions or different CM on postcontrast AKI may
4.9%, respectively. Using baseline eGFR as a predictor for provide information about the clinical relevance of AKI
risk for AKI, the same authors found no difference in AKI following contrast exposure. Some RCTs of prophylactic
rates between CECT and unenhanced CT patients strategies have found an increase in adverse clinical out-
(Table 1).51 The findings of the McDonald et al studies are comes in conjunction with a higher frequency of CIN but
different from the Davenport et al48,49 studies, which are confounded by the potential effect on mortality by the
found statistically increased rates of AKI in CECT patients prophylactic strategy itself. The CARE study was an RCT
compared with unenhanced CT patients with comparing an IOCM versus a LOCM with end points of
eGFR < 30 mL/min/1.73 m2 or Scr level ≥ 1.5 mg/dL and CIN rate and adverse clinical outcomes.69 This study
borderline statistical significance with eGFR of 30 to demonstrated a lower rate of CIN in the LOCM group
44 mL/min/1.73 m2. McDonald et al suggest that the compared to the IOCM group. Long-term follow-up
discrepancy may be explained by different propensity showed a lower rate of major adverse events (death,
methodologies and/or sample size of patients with stroke, myocardial infarction, or kidney failure) among
eGFR < 30 mL/min/1.73 m2. those who received LOCM as compared to IOCM, sup-
These same investigators have performed additional porting the possibility that CIN directly can contribute to
propensity score analyses in specific cohorts with high AKI these clinical outcomes.70
risk.52-54 The results of these studies did not show an If CM is nephrotoxic, adverse clinical outcomes from
increased risk for AKI after CM exposure, even after risk AKI following intra-arterial CM exposure would be rele-
stratification by eGFR (Table 1). Among a cohort exposed vant to AKI from intravenous CM exposure. Adverse events
to iso-osmolal contrast media (IOCM) only, there was a of AKI following intravenous CM exposure have been less
trend toward greater AKI in the unenhanced CT compared frequently studied compared to coronary angiography and
with CECT in individuals with eGFRs < 60 mL/min/ have demonstrated conflicting findings. A few studies have
1.73 m2, suggesting residual selection bias despite pro- shown increased risk for in-hospital, 30-day, and 1-year
pensity score matching.53 In an intensive care unit setting, mortality in individuals who developed AKI following
patients with eGFRs < 45 mL/min/1.73 m2 exposed to intravenous CM administration.71-74 However the validity
CECT had increased odds of emergent dialysis as compared of these results is questionable because in one particular
with those exposed to unenhanced CT (OR, 2.72; 95% CI, study, 12% of patients with AKI required renal replace-
1.14-6.46).54 Finally, Hinson et al55 performed a pro- ment therapy and 29% were oliguric, which is atypical of
pensity score analysis in a single-center emergency CIN.71 Other studies contradict these associations, showing
department setting that showed similar AKI in CECT and no difference in adverse long-term outcomes of eGFR
unenhanced CT patients irrespective of AKI definition or decline, 30-day mortality, or need for emergent dialysis
eGFR stratification. Baseline Scr level in the entire cohort following CECT exposure.75-78 When evaluating risk for
was 1.0 mg/dL and only 3.3% and 1.0% of patients had kidney failure in patients with CKD exposed to CECT and
eGFRs of 30 to 44 or <30 mL/min/1.73 m2, respectively. unenhanced CT, Hsieh et al78 found no difference among
the exposure groups, although in individuals with more
Mortality, Dialysis, and CKD With CIN frequent CECT per year, there was an increased risk for
kidney failure compared with patients with more frequent
AKI is associated with risks for CKD development, pro-
unenhanced CT exposure.78 Application of this conclusion
gression, and all-cause mortality.56-60 These observational
is questionable because underlying comorbid conditions
studies are supported by experimental studies of AKI that
requiring more frequent CECT studies may have also been
provide plausible biological mechanisms for clinically
responsible for the increased risk.
relevant outcomes.61-64 In a population that has developed
CIN, there is a debate of whether this is associated with a
clinically relevant longer-term adverse outcome. Several Summary and Recommendations
studies have demonstrated increased in-hospital and long- It is important to determine with as much precision as
term mortality, CKD progression, and kidney failure in possible the true risk for nephrotoxicity from intravenous
patients who developed AKI following contrast angiog- CM exposure. If the risk is overestimated, patients who
raphy, compared with those without AKI following cor- could clinically benefit from CECT would be deprived of
onary angiography.65-68 Of course, observational studies that benefit and there would be needless application of
do not prove a causal relationship between CIN and resources to prevent against this complication. If the risk is
adverse clinical outcomes because the association could be underestimated, patients could be exposed to a nephro-
due to confounding comorbid conditions that predispose toxic insult with the potential for adverse clinical out-
to both CIN and adverse clinical outcomes.65 The effect of comes, including progression of CKD.
non-CM confounding comorbid conditions on adverse Initial studies of AKI following intravenous CM were
clinical outcomes may be more relevant with intra-arterial uncontrolled and reported a wide range of AKI rates in a
than intravenous studies of CIN. variety of clinical settings. On average, the incidence of

AJKD Vol 75 | Iss 1 | January 2020 109


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

AKI following intravenous CM exposure was determined included in the propensity score, including prophylactic
to be 5% to 6%, leading many to conclude that AKI risk strategies and adjustment for peri-scan medications known
with intravenous CM was lower than AKI risk reported to affect Scr levels. Another major limitation of existing
after coronary angiography and thus intravenous CM propensity score studies is the low number of patients with
administration may be less nephrotoxic than intra-arterial severely decreased eGFRs who have the highest risk for
CM administration. However, differences in AKI rates CIN (Table 1). Furthermore, in none of these studies was a
following intravenous and intra-arterial CM administration separate propensity score analysis performed for patients
are more likely a function of comorbidity and procedural stratified by eGFR and DM together. Other limitations of
differences between the 2 patient populations. The litera- these propensity score studies include binary measurement
ture of CIN also contains numerous studies comparing a of confounding covariates, inclusion of patients with un-
CM-exposed group with a nonexposed and non- stable baseline Scr values, and analysis of only inpatients
randomized control group that consistently demonstrated with adequate Scr data, which predisposes to ascertain-
equivalent rates of AKI in both groups. However, these ment bias. 82
studies had numerous methodologic limitations that limit So how should physicians interpret the risk for AKI
this conclusion. These controlled studies were all obser- from intravenous CM exposure? Existing evidence points
vational, thus subject to selection bias, whereby physicians to a negligible risk for CIN following intravenous CM
may avoid CM exposure in patients believed to be at exposure in patients with eGFRs ≥ 60 mL/min/1.73 m2 or
highest risk for CIN due to other comorbid conditions.11 mildly decreased kidney function (eGFRs of 45-59 mL/
The presence of these unmeasured risk factors in the min/1.73 m2). It also seems clear that patients with
unenhanced CT controls was undoubtedly the cause of AKI eGFRs < 30 mL/min/1.73 m2 are at greatest risk for CIN
observed in these control groups. However, if these pre- following intravenous CM exposure, despite mixed find-
disposing AKI risk factors were present to a lesser degree in ings of the propensity score studies (Table 1). This leaves
the CECT patients, this would have the biased the effect of open the question on how to risk classify patients with
CM in the AKI toward no difference when compared with eGFRs of 30 to 44 mL/min/1.73 m2. Fortunately, this
unenhanced CT control groups.2,79 group of patients constitutes only a minority of patients
Despite these limitations, these studies led to 2 con- who undergo CT.83 Although guidelines by the European
clusions that are valid. First, previously reported rates of Society of Urogenital Radiology Contrast Medium Safety
AKI from intravenous CM are in general overstated. Sec- Committee and American College of Radiology suggest
ond, the occurrence of AKI following intravenous CM that this group is not at increased risk for CIN, in our
should not be automatically inferred as being due to CIN. opinion, there remains lingering concerns about this
We agree with the American College of Radiology conclusion.12,30 Our concern for CIN is further heightened
recommendation that the term postcontrast AKI instead of when patients with CKD in this eGFR range also have DM.
CIN should be used when the cause of AKI post–CM We suggest that risk classification for AKI following
exposure is unclear. intravenous CM exposure should not be based solely on a
Recognizing the limitations of these nonrandomized single eGFR determination. This is especially true for pa-
controlled studies, propensity score studies were per- tients with eGFRs of 30 to 45 mL/min/1.73 m2 and in
formed in an attempt to remove the imbalance of AKI- some patients with eGFRs < 30 mL/min/1.73 m2. We
predisposing comorbid conditions between the CM and recommend ensuring that the patient has a stable Scr level
control groups 80 All of the propensity scores have before CM exposure. Second, physicians should take into
demonstrated an equivalent rate of AKI after CM in patients consideration each patient’s unique risk factors for CIN
with normal or mildly decreased kidney function, which following intravenous CM exposure and weigh this against
was not surprising given the known absence of CIN in the clinical benefit of performing CECT. Third, we
these populations. However, in these propensity score encourage increased discussions between radiologists and
studies, the risk for CIN following CM exposure in patients physicians ordering CT on the added clinical value of CM
with moderate to severe CKD has been conflicting. Pro- enhancement. In patients at risk for CIN, noniodinated
pensity score methods are used in an attempt to minimize contrast studies including contrast-enhanced ultrasound or
selection bias by balancing measurable confounders; magnetic resonance imaging with newer gadolinium-
however, unmeasured confounders may still influence based contrast agents may provide adequate diagnostic
outcomes.81 The robust numbers and sophisticated information without a risk for nephrotoxicity.
matching techniques of these propensity score studies have If CM exposure is unavoidable in patients at high risk
led many to conclude that the risk for CIN is negligibly for CIN, we recommend prophylactic saline solution hy-
low or nonexistant and is clinically insignificant. Because dration and techniques to minimize contrast volume.
observational studies lack randomization, the patients Spacing of CM exposures remains a reasonable but un-
exposed to a treatment, in this case CM, are almost proven precaution. Although controlled studies of AKI
certainly different from those who are not exposed to the following intra-arterial CM exposure have not been done
treatment. The propensity score studies of intravenous for obvious reasons, our recommendations would also
CIN have one or more known confounders that are not apply to risk for AKI with intra-arterial CM exposure.

110 AJKD Vol 75 | Iss 1 | January 2020


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

Finally, we recommend that additional adequately pow- 4. Detrenis S, Meschi M, Musini S, Savazzi G. Lights and
ered propensity score studies should be conducted in pa- shadows on the pathogenesis of contrast-induced nephropa-
tients with eGFRs < 45 mL/min/1.73 m2 with and thy: state of the art. Nephrol Dial Transplant. 2005;20(8):1542-
1550.
without DM to further clarify the true risk for CIN in these 5. Heyman SN, Rosenberger C, Rosen S. Regional alterations in
presumed at-risk patients. renal haemodynamics and oxygenation: a role in contrast
medium-induced nephropathy. Nephrol Dial Transplant.
Review of the Clinical Vignette 2005;20(suppl 1):i6-i11.
6. Katzberg RW, Morris TW, Lasser EC, et al. Acute systemic and
The patient was suspected of having a pulmonary embo- renal hemodynamic effects of meglumine/sodium diatrizoate
lism and a decision was made to proceed with CTA with 76% and iopamidol in euvolemic and dehydrated dogs. Invest
intravenous CM. Because the patient was considered high Radiol. 1986;21(10):793-797.
risk for CIN, he received normal saline solution (240 mL 7. Persson PB, Tepel M. Contrast medium-induced nephropathy:
over 1 hour) before contrast exposure, which was the pathophysiology. Kidney Int. 2006;69(suppl 100):S8-S10.
8. Rudnick MR, Goldfarb S. Pathogenesis of contrast-induced
continued at 100 mL/h for 6 hours after the procedure.
nephropathy: experimental and clinical observations with an
CTA identified a segmental pulmonary embolism. The emphasis on the role of osmolality. Rev Cardiovasc Med.
following day, Scr level was 2.5 mg/dL with urine output 2003;4(suppl 5):S28-S33.
of 1,100 mL and then started to decline, returning to an 9. Katzberg RW, Newhouse JH. Intravenous contrast
Scr level of 1.9 mg/dL by the third day. medium-induced nephrotoxicity: is the medical risk really as
great as we have come to believe? Radiology.
2010;256(1):21-28.
Supplementary Material 10. McDonald RJ, McDonald JS, Newhouse JH, Davenport MS.
Supplementary File (PDF) Controversies in contrast material-induced acute kidney injury:
Table S1: Incidence of CIN and adverse outcomes following IV closing in on the truth? Radiology. 2015;277(3):627-632.
11. Newhouse JH, RoyChoudhury A. Quantitating contrast
contrast exposure (controlled studies).
medium-induced nephropathy: controlling the controls. Radi-
ology. 2013;267(1):4-8.
Article Information 12. American College of Radiology. ACR clinical appropriate-
Authors’ Full Names and Academic Degrees: Michael R. Rudnick, ness criteria. https://www.acr.org/Clinical-Resources/ACR-
MD, Amanda K. Leonberg-Yoo, MD, MS, Harold I. Litt, MD, Raphael Appropriateness-Criteria. Accessed January 31, 2019.
M. Cohen, MD, Susan Hilton, MD, and Peter P. Reese, MD, MSCE. 13. Rudnick MR, Goldfarb S, Wexler L, et al. Nephrotoxicity of ionic
Authors’ Affiliations: Divisions of Nephrology (MRR, AKL-Y, RMC, and nonionic contrast media in 1196 patients: a randomized
PPR) and Radiology (HIL, SH), Perelman School of Medicine at trial. The Iohexol Cooperative Study. Kidney Int. 1995;47(1):
the University of Pennsylvania, Philadelphia, PA. 254-261.
Address for Correspondence: Michael R. Rudnick, MD, Perelman 14. Eng J, Wilson RF, Subramaniam RM, et al. Comparative effect
School of Medicine at the University of Pennsylvania, 51 N 39th of contrast media type on the incidence of contrast-induced
St, Medical Office Bldg, Ste 240, Philadelphia, PA 19104. E-mail: nephropathy: a systematic review and meta-analysis. Ann
michael.rudnick@uphs.upenn.edu Intern Med. 2016;164(6):417-424.
15. Heinrich MC, Haberle L, Muller V, Bautz W, Uder M. Nephro-
Support: There was no additional support provided for the work
described in this article. toxicity of iso-osmolar iodixanol compared with nonionic
low-osmolar contrast media: meta-analysis of randomized
Financial Disclosure: The authors declare that they have no controlled trials. Radiology. 2009;250(1):68-86.
relevant financial interests.
16. Weisbord SD, Palevsky PM. Prevention of contrast-induced
Other Disclosures: Dr Reese serves as an AJKD Associate Editor; nephropathy with volume expansion. Clin J Am Soc Nephrol.
he was entirely recused from the manuscript consideration process. 2008;3(1):273-280.
Peer Review: Received November 12, 2018, in response to an 17. Jurado-Roman A, Hernandez-Hernandez F, Garcia-Tejada J,
invitation from the journal. Evaluated by 2 external peer reviewers et al. Role of hydration in contrast-induced nephropathy in
and a member of the Feature Advisory Board, with direct editorial patients who underwent primary percutaneous coronary inter-
input from the Feature Editor and a Deputy Editor. Accepted in vention. Am J Cardiol. 2015;115(9):1174-1178.
revised form May 7, 2019. 18. Luo Y, Wang X, Ye Z, et al. Remedial hydration reduces the
incidence of contrast-induced nephropathy and short-term
adverse events in patients with ST-segment elevation myocar-
References dial infarction: a single-center, randomized trial. Intern Med.
1. Bartels ED, Brun GC, Gammeltoft A, Gjørup PA. Acute anuria 2014;53(20):2265-2272.
following intravenous pyelography in a patient with myeloma- 19. Mueller C, Buerkle G, Buettner HJ, et al. Prevention of contrast
tosis. Acta Med Scand. 1954;150(4):297-302. media-associated nephropathy: randomized comparison of 2
2. Luk L, Steinman J, Newhouse JH. Intravenous contrast-induced hydration regimens in 1620 patients undergoing coronary an-
nephropathy-the rise and fall of a threatening idea. Adv Chronic gioplasty. Arch Intern Med. 2002;162(3):329-336.
Kidney Dis. 2017;24(3):169-175. 20. Shah R, Wood SJ, Khan SA, Chaudhry A, Rehan Khan M,
3. Parfrey PS, Griffiths SM, Barrett BJ, et al. Contrast material- Morsy MS. High-volume forced diuresis with matched hydration
induced renal failure in patients with diabetes mellitus, renal using the RenalGuard System to prevent contrast-induced
insufficiency, or both. A prospective controlled study. N Engl J nephropathy: a meta-analysis of randomized trials. Clin Car-
Med. 1989;320(3):143-149. diol. 2017;40(12):1242-1246.

AJKD Vol 75 | Iss 1 | January 2020 111


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

21. Ramirez G, O’Neill WM Jr, Lambert R, Bloomer HA. Cholesterol 38. Moos SI, van Vemde DN, Stoker J, Bipat S. Contrast induced
embolization: a complication of angiography. Arch Intern Med. nephropathy in patients undergoing intravenous (IV) contrast
1978;138(9):1430-1432. enhanced computed tomography (CECT) and the relationship
22. Feldkamp T, Luedemann M, Spehlmann ME, et al. Radial ac- with risk factors: a meta-analysis. Eur J Radiol. 2013;82(9):
cess protects from contrast media induced nephropathy after e387-e399.
cardiac catheterization procedures. Clin Res Cardiol. 39. Newhouse JH, Kho D, Rao QA, Starren J. Frequency of serum
2018;107(2):148-157. creatinine changes in the absence of iodinated contrast ma-
23. Katzberg RW, Lamba R. Contrast-induced nephropathy after terial: implications for studies of contrast nephrotoxicity. AJR
intravenous administration: fact or fiction? Radiol Clin North Am J Roentgenol. 2008;191(2):376-382.
Am. 2009;47(5):789-800, v. 40. Bruce RJ, Djamali A, Shinki K, Michel SJ, Fine JP, Pozniak MA.
24. Pandya B, Chalhoub JM, Parikh V, et al. Contrast media use in Background fluctuation of kidney function versus contrast-
patients with chronic kidney disease undergoing coronary induced nephrotoxicity. AJR. Am J Roentgenol. 2009;192(3):
angiography: a systematic review and meta-analysis of ran- 711-718.
domized trials. Int J Cardiol. 2017;228:137-144. 41. Khwaja A. KDIGO clinical practice guidelines for acute kidney
25. Nyman U, Almen T, Jacobsson B, Aspelin P. Are intravenous injury. Nephron Clin Pract. 2012;120(4):c179-c184.
injections of contrast media really less nephrotoxic than intra- 42. Aycock RD, Westafer LM, Boxen JL, Majlesi N, Schoenfeld EM,
arterial injections? Eur Radiol. 2012;22(6):1366-1371. Bannuru RR. Acute kidney injury after computed tomography: a
26. Stratta P, Izzo C, Canavese C, Quaglia M. Letter to the Editor. meta-analysis. Ann Emerg Med. 2018;71(1):44-53.
Are intravenous injections of contrast media really less neph- 43. Wilhelm-Leen E, Montez-Rath ME, Chertow G. Estimating the
rotoxic than intra-arterial injections? Eur Radiol. 2013;23: risk of radiocontrast-associated nephropathy. J Am Soc
1260-1263. Nephrol. 2017;28(2):653-659.
27. Stacul F, van der Molen AJ, Reimer P, et al. on behalf of the 44. McDonald JS, McDonald RJ, Comin J, et al. Frequency of acute
Contrast Media Safety Coommittee of European Society of kidney injury following intravenous contrast medium adminis-
Urogenital Radiology (ESUR). Contrast induced nephropathy: tration: a systematic review and meta-analysis. Radiology.
updated ESUR contrast media safety guidelines. Eur Radiol. 2013;267(1):119-128.
2011;21:2527-2541. 45. Kayibanda JF, Hiremath S, Knoll GA, et al. Does intravenous
28. Davidson C, Stacul F, McCullough PA, et al; on behalf of the contrast-enhanced computed tomography cause acute kidney
CIN Consensus Working Panel. Contrast medium use. Am J injury? Protocol of a systematic review of the evidence. Syst
Cardiol. 2006;98(suppl 1):42K-58K. Rev. 2014;3:94.
29. Gutierrez NM, Newhouse JH. Maximum arterial contrast con- 46. Kooiman J, Pasha SM, Zondag W, et al. Meta-analysis: serum
centrations with computed tomography and left ventriculog- creatinine changes following contrast enhanced CT imaging.
raphy: implications for contrast nephrotoxicity risk. J Comput Eur J Radiol. 2012;81(10):2554-2561.
Assist Tomogr. 2017;41(6):976-982. 47. Moore A, Dickerson E, Dillman JR, Vummidi D, Kershaw DB.
30. van der Molen AJ, Reimer P, Dekkers IA, et al. Post-contrast Incidence of nonconfounded post–computed tomography
acute kidney injury. Part 2: risk stratification, role of hydration acute kidney injury in hospitalized patients with stable renal
and other prophylactic measures, patients taking metformin function receiving intravenous iodinate. Curr Probl Diagn
and chronic dialysis patients: recommendations for updated Radiol. 2014;43(5):237-241.
ESUR Contrast Medium Safety Committee guidelines. Eur 48. Davenport MS, Khalatbari S, Dillman JR, Cohan RH, Caoili EM,
Radiol. 2018;28:2856-2869. Ellis JH. Contrast material-induced nephrotoxicity and intrave-
31. Karlsberg RP, Dohad SY, Sheng R. Contrast medium-induced nous low-osmolality iodinated contrast material. Radiology.
acute kidney injury: comparison of intravenous and intra- 2013;267(1):94-105.
arterial administration of iodinated contrast medium. J Vasc 49. Davenport MS, Khalatbari S, Cohan RH, Dillman JR, Myles JD,
Interv Radiol. 2011;22(8):1159-1165. Ellis JH. Contrast material-induced nephrotoxicity and intrave-
32. Kooiman J, Le Haen PA, Gezgin G, et al. Contrast-induced nous low-osmolality iodinated contrast material: risk stratifica-
acute kidney injury and clinical outcomes after intra-arterial and tion by using estimated glomerular filtration rate. Radiology.
intravenous contrast administration: risk comparison adjusted 2013;268(3):719-728.
for patient characteristics by design. Am Heart J. 2013;165(5): 50. McDonald RJ, McDonald JS, Bida JP, et al. Intravenous
793-799. 799.e791. contrast material-induced nephropathy: causal or coincident
33. McDonald JS, Leake CB, McDonald RJ, et al. Acute kidney phenomenon? Radiology. 2013;267(1):106-118.
injury after intravenous versus intra-arterial contrast material 51. McDonald JS, McDonald RJ, Carter RE, Katzberg RW,
administration in a paired cohort. Invest Radiol. 2016;51(12): Kallmes DF, Williamson EE. Risk of intravenous contrast
804-809. material-mediated acute kidney injury: a propensity score-
34. Cramer BC, Parfrey PS, Hutchinson TA, et al. Renal func- matched study stratified by baseline-estimated glomerular
tion following infusion of radiologic contrast material. A filtration rate. Radiology. 2014;271(1):65-73.
prospective controlled study. Arch Intern Med. 52. McDonald JS, McDonald RJ, Lieske JC, et al. Risk of acute
1985;145(1):87-89. kidney injury, dialysis, and mortality in patients with chronic
35. Rao QA, Newhouse JH. Risk of nephropathy after intravenous kidney disease after intravenous contrast material exposure.
administration of contrast material: a critical literature analysis. Mayo Clin Proc. 2015;90(8):1046-1053.
Radiology. 2006;239(2):392-397. 53. McDonald JS, McDonald RJ, Williamson EE, Kallmes DF. Is
36. Katzberg RW, Barrett BJ. Risk of iodinated contrast material– intravenous administration of iodixanol associated with
induced nephropathy with intravenous administration. Radi- increased risk of acute kidney injury, dialysis, or mortality? A
ology. 2007;243(3):622-628. propensity score-adjusted study. Radiology. 2017;285(2):414-
37. Heller CA, Knapp J, Halliday J, O’Connell D, Heller RF. Failure 424.
to demonstrate contrast nephrotoxicity. Med J Aust. 54. McDonald JS, McDonald RJ, Williamson EE, Kallmes DF,
1991;155(5):329-332. Kashani K. Post-contrast acute kidney injury in intensive care

112 AJKD Vol 75 | Iss 1 | January 2020


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
In Practice

unit patients: a propensity score-adjusted study. Intensive Care 70. Solomon RJ, Mehran R, Natarajan MK, et al. Contrast-induced
Med. 2017;43(6):774-784. nephropathy and long-term adverse events: cause and effect?
55. Hinson JS, Ehmann MR, Fine DM, et al. Risk of acute kidney Clin J Am Soc Nephrol. 2009;4(7):1162-1169.
injury after intravenous contrast media administration. Ann 71. Levy EM, Viscoli CM, Horwitz RI. The effect of acute renal
Emerg Med. 2017;69(5):577-586. failure on mortality. A cohort analysis. JAMA. 1996;275(19):
56. Coca SG, Peixoto AJ, Garg AX, Krumholz HM, Parikh CR. The 1489-1494.
prognostic importance of a small acute decrement in kidney 72. From AM, Bartholmai BJ, Williams AW, Cha SS, McDonald FS.
function in hospitalized patients: a systematic review and meta- Mortality associated with nephropathy after radiographic
analysis. Am J Kidney Dis. 2007;50(5):712-720. contrast exposure. Mayo Clin Proc. 2008;83(10):1095-1100.
57. Coca SG, Yusuf B, Shlipak MG, Garg AX, Parikh CR. Long- 73. Mitchell AM, Jones AE, Tumlin JA, Kline JA. Incidence of
term risk of mortality and other adverse outcomes after acute contrast-induced nephropathy after contrast-enhanced
kidney injury: a systematic review and meta-analysis. Am J computed tomography in the outpatient setting. Clin J Am
Kidney Dis. 2009;53(6):961-973. Soc Nephrol. 2010;5(1):4-9.
58. Singbartl K, Kellum JA. AKI in the ICU: definition, epidemiology, 74. Mitchell AM, Kline JA, Jones AE, Tumlin JA. Major adverse
risk stratification, and outcomes. Kidney Int. 2012;81(9):819- events one year after acute kidney injury after contrast-
825. enhanced computed tomography. Ann Emerg Med.
59. Shiao CC, Wu PC, Huang TM, Lai TS, Yang WS. Long-term 2015;66(3):267-274.
remote organ consequences following acute kidney injury. Crit 75. Weisbord SD, Mor MK, Resnick AL, Hartwig KC, Palevsky PM,
Care. 2015;19(1):1-11. Fine MJ. Incidence and outcomes of contrast-induced AKI
60. Wang HE, Muntner P, Chertow GM, Warnock DG. Acute kid- following computed tomography. Clin J Am Soc Nephrol.
ney injury and mortality in hospitalized patients. Am J Nephrol. 2008;3(5):1274-1281.
2012;35(4):349-355. 76. McDonald RJ, McDonald JS, Carter RE, et al. Intravenous
61. Parr SK, Siew ED. Delayed consequences of acute kidney contrast material exposure is not an independent risk factor for
injury. Adv Chronic Kidney Dis. 2016;23(3):186-194. dialysis or mortality. Radiology. 2014;273(3):714-725.
62. Depret F, Prud’homme M, Legrand M. A role of remote organs 77. Kim IY, Lee SB, Lee DW, Song SH, Seong EY, Kwak IS. Long-
effect in acute kidney injury outcome. Nephron. 2017;137(4): term effect of radiocontrast-enhanced computed tomography
273-276. on the renal function of chronic kidney disease patients. Clin
63. Ologunde R, Zhao H, Lu K, Ma D. Organ cross talk and remote Exp Nephrol. 2012;16(5):755-759.
organ damage following acute kidney injury. Int Urol Nephrol. 78. Hsieh MS, Chiu CS, How CK, et al. Contrast medium exposure
2014;46(12):2337-2345. during computed tomography and risk of development of end-
64. Basile DP, Friedrich JL, Spahic J, et al. Impaired endothelial stage renal disease in patients with chronic kidney disease: a
proliferation and mesenchymal transition contribute to vascular nationwide population-based, propensity score-matched, lon-
rarefaction following acute kidney injury. Am J Physiol Renal gitudinal follow-up study. Medicine. 2016;95(16):e3388.
Physiol. 2010;300:F721-F733. 79. Davenport MS, Cohan RH, Khalatbari S, Ellis JH. The chal-
65. Rudnick M, Feldman H. Contrast-induced nephropathy: what lenges in assessing contrast-induced nephropathy: where are
are the true clinical consequences? Clin J Am Soc Nephrol. we now? AJR Am J Roentgenol. 2014;202(4):784-789.
2008;3(1):263-272. 80. Dekkers IA, van der Molen AJ. Propensity score matching as a
66. Maioli M, Toso A, Leoncini M, Gallopin M, Musilli N, Bellandi F. substitute for randomized controlled trials on acute kidney injury
Persistent renal damage after contrast-induced acute kidney after contrast media administration. A systematic review. AJR
injury. Circulation. 2012;125:3099-3107. Am J Roentgenol. 2018;211(4):822-826.
67. Goldenberg I, Chonchol M, Guetta V. Reversible acute kidney 81. Austin PC. An introduction to propensity score methods for
injury following contrast exposure and the risk of long-term reducing the effects of confounding in observational studies.
mortality. Am J Nephrol. 2009;29:136-144. Multivariate Behav Res. 2011;46(3):399-424.
68. James MT, Samuel SM, Manning MA, et al. Contrast-induced 82. Nyman U, Aspelin P, Jakobsen J, Bjork J. Controversies in
acute kidney injury and risk of adverse clinical outocomes after contrast material- induced acute kidney injury: propensity score
coronary angiography. A systematic review and meta-analysis. matching of patients with different dose/absolute glomerular
Circ Cardiovasc Interv. 2013;6:37-43. filtration ratios. Radiology. 2015;277(3):633-637.
69. Solomon RJ, Natarajan MK, Doucet S, et al. Cardiac Angiog- 83. Herts BR, Schneider E, Poggio ED, Obuchowski NA,
raphy in Renally Impaired Patients (CARE) study: a randomized Baker ME. Identifying outpatients with renal insufficiency before
double-blind trial of contrast-induced nephropathy in patients contrast-enhanced CT by using estimated glomerular filtration
with chronic kidney disease. Circulation. 2007;115(25):3189- rates versus serum creatinine levels. Radiology. 2008;248(1):
3196. 106-113.

AJKD Vol 75 | Iss 1 | January 2020 113


Descargado para Anonymous User (n/a) en Cayetano Heredia Pervuvian University de ClinicalKey.es por Elsevier en abril 28, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.

You might also like