Advances in Cardiac Disease

Download as pdf or txt
Download as pdf or txt
You are on page 1of 390
At a glance
Powered by AI
The document discusses topics related to coronary atherosclerosis and advances in its diagnosis, including mechanisms of disease, diagnostic techniques like coronary angiography, and risks/complications.

The book discusses various topics related to coronary atherosclerosis, its diagnosis using techniques like coronary angiography, and risks/complications of such diagnostic procedures.

Some methods discussed to reduce radiation exposure include reducing patient exposure, increasing distance from the radiation source, and using shielding.

ADVANCES IN THE

DIAGNOSIS OF CORONARY
ATHEROSCLEROSIS
Edited by Suna F. Kiraç
Advances in the Diagnosis of Coronary Atherosclerosis
Edited by Suna F. Kiraç

Published by InTech
Janeza Trdine 9, 51000 Rijeka, Croatia

Copyright © 2011 InTech


All chapters are Open Access distributed under the Creative Commons Attribution 3.0
license, which permits to copy, distribute, transmit, and adapt the work in any medium,
so long as the original work is properly cited. After this work has been published by
InTech, authors have the right to republish it, in whole or part, in any publication of
which they are the author, and to make other personal use of the work. Any republication,
referencing or personal use of the work must explicitly identify the original source.

As for readers, this license allows users to download, copy and build upon published
chapters even for commercial purposes, as long as the author and publisher are properly
credited, which ensures maximum dissemination and a wider impact of our publications.

Notice
Statements and opinions expressed in the chapters are these of the individual contributors
and not necessarily those of the editors or publisher. No responsibility is accepted for the
accuracy of information contained in the published chapters. The publisher assumes no
responsibility for any damage or injury to persons or property arising out of the use of any
materials, instructions, methods or ideas contained in the book.

Publishing Process Manager Sandra Bakic


Technical Editor Teodora Smiljanic
Cover Designer Jan Hyrat
Image Copyright Lightspring, 2011. Used under license from Shutterstock.com

First published October, 2011


Printed in Croatia

A free online edition of this book is available at www.intechopen.com


Additional hard copies can be obtained from [email protected]

Advances in the Diagnosis of Coronary Atherosclerosis, Edited by Suna F. Kiraç


p. cm.
ISBN 978-953-307-286-9
free online editions of InTech
Books and Journals can be found at
www.intechopen.com
Contents

Preface IX

Chapter 1 Mechanisms of Disease:


Novel Polymorphisms in Coronary Artery Disease 1
Asghar Ghasemi, Morteza Seifi and Mahmood Khosravi

Chapter 2 Multifunctional Role of TRAIL in Atherosclerosis


and Cardiovascular Disease 19
Katsuhito Mori, Masanori Emoto and Masaaki Inaba

Chapter 3 Indications for Coronary Angiography 33


Karl Poon and Darren Walters

Chapter 4 History of Coronary Angiography 69


Ryotaro Wake, Minoru Yoshiyama, Hidetaka Iida,
Hiroaki Takeshita, Takanori Kusuyama,
Hitoshi Kanamitsu, Hideya Mitsui, Yukio Yamada,
Shinichi Shimodozono and Kazuo Haze

Chapter 5 Coronary Angiography -


Physical and Technical Aspects 81
Maria Anna Staniszewska

Chapter 6 Procedural Techniques of Coronary Angiography 95


Jasmin Čaluk

Chapter 7 Risks and Complications of Coronary Angiography:


Contrast Related Complications 121
S. Mohammad Reza Khatami

Chapter 8 Complications of Cardiac Catetherization 149


Mariano García-Borbolla, Rafael García-Borbolla
and Begoña Balboa

Chapter 9 Diagnosis and Management of Complications


of Invasive Coronary Angiography 169
Jong-Seon Park and Young-Jo Kim
VI Contents

Chapter 10 Coronary Angiography


and Contrast-Induced Nephropathy 181
Omer Toprak

Chapter 11 Coronary Angiography in Patients


with Chronic Kidney Disease 203
Luís Henrique Wolff Gowdak and José Jayme Galvão de Lima

Chapter 12 Cardiac Catheterization and Coronary Angiography in


Patients with Cardiomyopathy 219
Ali Ghaemian

Chapter 13 Contrast-Induced Nephropathy in Patients with Type 2


Diabetes Mellitus and Coronary Artery Disease:
Update and Practical Clinical Applications 235
Richard E. Katholi and Charles R. Katholi

Chapter 14 Quantitative Coronary Angiography


in the Interventional Cardiology 255
Salvatore Davide Tomasello, Luca Costanzo
and Alfredo Ruggero Galassi

Chapter 15 Summarized Coronary Artery Caliber and Left


Ventricle Mass for Scoring of Cardiac Ischemia:
Diagnostic and Prognostic Value 273
Edvardas Vaicekavicius

Chapter 16 Woven Coronary Artery 297


Ayşe Yıldırım and A. Deniz Oğuz

Chapter 17 Image Post-Processing and Interpretation 305


Masahiro Jinzaki, Minoru Yamada and Sachio Kuribayashi

Chapter 18 Novel Insights Into Stenosis on Coronary


Angiography–Outline of Functional Assessment
of Stable Angina Patients with Angiographic Stenosis 331
Shinichiro Tanaka

Chapter 19 Optimization of Radiation Dose and Image Quality


in Cardiac Catheterization Laboratories 345
Octavian Dragusin, Christina Bokou,
Daniel Wagner and Jean Beissel

Chapter 20 Protection of the Patient and the Staff from


Radiation Exposure During Fluoroscopy-Guided
Procedures in Cardiology 367
Verdun Francis R., Aroua Abbas, Samara Eleni,
Bochud François and Stauffer Jean-François
Preface

Coronary artery disease (CAD) and its consequences are the most important morbidity
and mortality reasons in the developed and developing countries. Advanced imaging
techniques (intravascular ultrasound, MR and CT angiography, SPECT/CT, PET/CT,
PET/MRI) and novel serologic biomarkers (C-reactive protein, interleukin 6, matrix
metalloproteinase, P-selectin, intracellular adhesion molecule 1 and tumor necrosis
factor ) provide early diagnosis of CAD and protect patients from hard cardiac
events. Non-invasive techniques are being widely used in the diagnosis and
management while conventional CAG is still the most commonly performed test in the
cases at high risk. Following the first cardiac catheterization performed, first selective
CAG has been reported at the end of 1950's. Patient specific and procedure-related
complications range widely from minor ones with short term sequelae to life
threatening events that may cause irreversible end-point if urgent treatment is not
adequately provided. The important risk factors for complications are older age, renal
insufficiency, uncontrolled diabetes mellitus, morbid obesity, and iodine allergy.
However, operator skills and the type of invasive procedure being performed remain
as the most important predictors to undesired outcomes. The risk-to-benefit ratio of
the CAG should be considered carefully on an individual basis.

Coronary CTA and CMRA among advanced imaging systems offer anatomical
informations not only for coronary vessels but also for peripheral vascular structures,
and assessment of the left and right ventricular functions is possible in same image
series. Quantified coronary artery calcification and many post-processing images (2-D
images and the different 3-D rendering images such as volume rendering, multiplanar
reformation, partial maximum intensity projection, curved multiplanar reformation)
should be evaluated to increase diagnostic accuracy. High calcification level signs
atherosclerotic changes in the coronary arteries, but is not specific for luminal
obstruction. Because the absence of detectable calcium deposition has a high negative
predictive value for CAD, CAC value is a significant predictive determinant for
prognosis in asymptomatic patients. As with coronary angiography, myocardial
perfusion abnormality may not be detected even there is coronary lesion causing a
luminal narrowing of greater than 50 % defined by CTA and MRA. In asymptomatic
and intermediate likelihood patients, assessment of myocardial perfusion by single
photon emission computed tomography (SPECT) or positron emission tomography
(PET) appears to be valuable even when coronary arteries are normal in angiography.
X Preface

If gated study is added, left ventricular systolic and diastolic functions can be
investigated simultaneously with myocardial perfusion. This field includes a overview
of molecular targeted imaging, permeability of the coronary vessel wall, and
interventional coronary MR. Recent developments in the field of ultrasonography
have allowed us to objectively quantify global and regional ventricular function, and
also, to get real-time evaluation of coronary walls and calcium load of atherosclerotic
plaques. While we achieve more knowledge about atherosclerotic lesions by IVUS,
tissue Doppler imaging has attempted us to assess myocardial function.

On the other hand, radiation exposure is the most limited factor for CTA and MPS
gated SPECT procedures and needs particular attention. Ionizing radiation doses,
hazardous effects and general radiation protection principles should be known for
optimal protection of the patients. Mainly radiation safety rules, various techniques
and equipments that may be used to reduce patient and staff radiation exposure
during diagnostic and therapeutic procedures especially cardiac interventional
fluoroscopic procedures have been detailed discussed in this book. In this field cardiac
MR, which is a powerful non-invasive technique for the simultaneously assessment of
coronary artery anatomy and function, has a great promise as a radiation-free method.
But, it currently lays behind CTA for noninvasive coronary angiography because of
some limitation factors such as metallic implants and equipment design.

Selection of the most appropriate diagnostic test in special situations such as chronic
kidney disease (CKD) and diabetes mellitus is an other important issue. Although
coronary angiography is a valuable tool, the major challenges with coronary
angiography relate to when it is appropriate to perform and what the risks are
associated with the procedure. Because renal function may be more and more impair
with contrast agents used during CAG, and sometimes dialysis may be needed.
Therefore, stress echocardiography, MRA and nuclear cardiac tests are often
recommended to rule out the presence of CAD in those patients and the presence of
any risk factor must be assessed on an individual basis in order to prevent for a soft or
hard local or systemic complications. Contrast induced nephropathy (CIN) remains an
important clinical issue in these patients, pre-treatment with theophylline combined
with volume expansion using sodium bicarbonate; acetylcysteine; use of the lowest
possible dose of contrast material (CM), and ISO-osmolar CM or low osmolar CM are
advised to prevent CIN. Contrast induced nephropathy is diagnosed if a rapid renal
dysfunction is occurred after CM administration without obviously any other cause of
acute kidney insufficiency. Serum creatinine (sCr) is the standard marker for detecting
CIN; however little changes in sCr after CM exposure may be seen but it is not
considered clinically relevant. Therefore, glomerular filtration rate which usually
measured by creatinine clearance is usually accepted as the most accurate method for
the assessment of kidney function. But, even in patients with stable sCr the GFR may
significantly be declined. Recently more sensitive markers (Cystatin-C and
Neutrophilic gelatinase associated lipocaline) than sCr for GFR have been developed
and validated. Cystatin-C is presented as more accurate marker than sCr for
predicting renal function. Readers will get detailed discussions about advantages,
Preface XI

disadvantages and possible complications of invasive and non-invasive cardiac


procedures, and test selection criteria based on patient’s characterizations, mechanism
and definition of contrast induced nephropathy (CIN) and preventive therapy models
in one more chapters of this task.

Consequently, this book summarized the clinics of atherosclerotic heart diseases,


pathogenesis covering possible genetic factors and risk factors, a current view on new
biomarkers as a diagnostic, prognostic parameters and future complications, novel
diagnostic imaging modalities for CAD, and their advantages and disadvantages.
Insight to molecular basis of CAD in a special chapter focusing on the role of on
TRAIL (Tumor necrosis factor (TNF)-related apoptosis-inducing ligand) in the
cardiovascular disease is really interesting and useful to understand how
atherosclerotic plaques occur and what the importance of administration of
recombinant TRAIL in protective therapy as a powerful approach.

Suna F. Kıraç
Pamukkale University, Faculty of Medicine, Denizli
Turkey
1

Mechanisms of Disease:
Novel Polymorphisms in
Coronary Artery Disease
Asghar Ghasemi1, Morteza Seifi2 and Mahmood Khosravi3
1Tabriz Health Center
2Department of Iranian Legal Medicine Organization
3Hematology Department of Medicine Faculty,

Ahvaz University of Medical Sciences


Iran

1. Introduction
Coronary artery disease (CAD) is one of the most common cardiovascular diseases and has
a high incidence of morbidity and mortality. CAD is a major public health problem in
developing and developed countries and its increasing prevalence is a cause of considerable
concern in the medical community worldwide (He et al., 2005). CAD involves genetic and
environmental factors and their interaction with each other. Traditional risk factors account
for at most one-half of the prevalence of CAD (Zdravkovic et al., 2002). Despite attempts to
establish the molecular and genetic determinants that could account for variations in CAD
(Zdravkovic et al., 2002), the etiology and complex multigenic basis of atherosclerosis is still
not completely understood.
Completion of the sequencing of the human genome was a monumental achievement (Venter
et al., 2001). Molecular researchers now take for granted the information provided by the
sequence, however the clinical applications are not immediately obvious. A limitation of the
Human Genome Project was that it produced only a single “reference” sequence. But in order
to identify new disease causing mechanisms and cures for disease, we need to go beyond the
“reference” and characterize the differences between our genomes, and in turn the effect that
these differences have. The Human HapMap consortium (Frazer et al., 2007) and recent
genome-wide association studies (GWAS) have set out to capture the interindividual
differences that are associated with disease processes, including coronary artery disease.
The association between genetic variations and CAD have been reviewed in several
previous manuscripts (Lanktree et al., 2008), but in our knowledge so far there has been no
study in the field of association between novel gene variations and CAD; therefore we
focused on introducing some novel polymorphisms and their relationships with CAD.

2. Genetics of coronary artery disease


2.1 Genetic architecture of CAD
The success of CAD gene mapping is dependent on its genetic architecture which refers to
the number of disease genes that exist, their allele frequencies, the risks that they confer, and
2 Advances in the Diagnosis of Coronary Atherosclerosis

the interactions between multiple genetic and environmental factors (Wright & Hastie, 2001;
Reich & Lander, 2001). Although the total genetic contribution to CAD risk can be
quantified, the determination of the size and number of contributing effects is impossible
without identifying all CAD susceptibility genes. The multiple risk factors for CAD
themselves have their own genetic architecture. The heritabilities of some of the risk factors
for CAD are considerable - total cholesterol (40 to 60%), HDL-cholesterol (45 to 75%), total
triglycerides (40 to 80%), body mass index (25 to 60%), systolic blood pressure (50 to 70%),
Lp(a) levels (90%), homocysteine levels (45%), type 2 diabetes (40 to 80%), fibrinogen (20 to
50%) (Lusis et al., 2004). Also, as CAD is rare before the age of 50 yr, it is unlikely to have an
effect on reproductive success and hence less likely to have been subject to direct
evolutionary selection pressure. Variants that confer susceptibility or protection for CAD
might therefore have evolved neutrally in the past, and so could present at a wide range of
frequencies.
This is the basis of the Common Disease/Common Variant (CDCV) hypothesis which holds
that the genetic variants underlying complex traits occur with a relatively high frequency
(>1%), have undergone little or no selection in earlier populations and are likely to date back
to >100,000 years ago (Lander et al., 1996).The other competing model is the Common
Disease Rare Variant hypothesis, with an inverse relationship between the magnitude of
genetic effect and allele frequency (Pritchard et al., 2001). This model argues that diseases
are common because of highly prevalent environmental influences, not because of common
disease alleles in the population (Wright & Hastie, 2001). A review of candidate gene
associations and recent genome wide association study results support the importance of
common alleles in CAD. At odds with this, rare allelic variants of three candidate genes
(ABCA1, APOA1, LCAT) that influence HDL levels, were jointly found to make a
substantial contribution to the population distribution of HDL levels (Cohen et al., 2004;
Frikke-Schmidt et al., 2004). The most likely scenario would be that the allelic spectrum of
the disease variants is the same as the general spectrum of all disease variants. Under this
neutral model, although most susceptibility variants are rare with minor allele frequencies
(MAF) <1 per cent, variants with MAF>1 per cent would account for more than 90 per cent
of the genetic differences between individuals. It is plausible that these common variants
might contribute significantly to those common diseases in which susceptibility alleles
might not be under intense negative selection.

2.2 Linkage and association studies


The 2 general types of studies that evaluate the relation between gene polymorphisms and
disease are linkage analysis and association studies. Linkage analysis investigates the
cosegregation of polymorphic DNA markers with inheritance of disease in families and has
been highly successful in the detection of monogenic disorders. However, it is a tedious and
complicated undertaking in the investigation of polygenic diseases such as CHD.
Association studies provide an alternative method for dissecting genetically complex
diseases and typically use the candidate gene approach for their investigation. Based on the
known pathophysiologic characteristics of a disease, assumptions are made about the genes
involved in its processes and the hypothesis of the association of these genes with the
disease is then tested. For a disease such as CHD it makes sense to analyze genes that
contribute to lipoprotein metabolism, blood pressure, and to diseases such as diabetes
mellitus, among others.
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 3

This approach is more directed than is the genome-scan linkage approach, but it is limited
by our incomplete knowledge of disease mechanisms and thus may miss important
causative genes. It is worth noting that whereas in linkage analyses “disease” alleles are
tracked in families, genetic association is a phenomenon of populations and association
studies compare populations of subjects with and without the disease of interest (Bernhard
et al., 2000).

2.3 Linkage studies


Previous methods for determining genetic linkage of a complex disease relied chiefly upon
the technique of genome wide scanning of microsatellites (short tandem repeat sequences).
This required the laborious selection of hundreds of families, particularly sib pairs with MI
or CAD. Using around 400 microsatellite markers distributed evenly across the genome, the
goal was to identify a significant linkage peak defined by a logarithm of odds ratio (LOD)
greater than 3.5 corresponding to a p<10-6 indicating a gene that is in linkage disequilibrium
(LD) near or even within the microsatellite region. However, identifying disease causing
genes interspersed within microsatellites has proved to be quite difficult and studies from
different centers reported varying results - Finland (2q21.2-22 and Xq23-26) (Pajukanta et al.
2000), Germany (14q32.2) (Broeckel et al., 2002), Iceland (13q12-13) ( Helgadottir et al., 2004),
US (1p34-36 ,3q13 and 5q31)( Hauser et al., 2004) and UK (2p11, 17p11-17q21) (Farrall et al.,
2006; Samani et al. 2005). Genomic regions identified in the published linkage studies as
being correlated with CHF are largely non-overlapping, suggesting genetic and/or
phenotypic heterogeneity. Two genes, ALOX5AP and MEF2A, have been identified by fine
mapping studies following the original linkage analysis. The Icelandic locus was replicated
in population-based studies from Iceland and England, with different haplotypes of the
ALOX5AP gene (encoding 5-lipoxygenase activating protein) associated with CAD in the
two countries, and the Icelandic haplotype was also associated with stroke in Iceland and in
Scotland (Helgadottir et al., 2004; Helgadottir et al., 2005). MEF2A (myocyte enhancer factor
2A) a transcription factor expressed in coronary artery endothelium was identified by
linkage analysis in a pedigree in which 13 members had CAD, nine of whom had MI (Wang
et al., 2003).

2.4 Genome-wide association studies


Genome-wide association studies (GWAS) became possible after the publication of the
International Haplotype Map Project (HapMap) (International HapMap consortium, 2005;
International HapMap consortium, 2007) and the development of array-based platforms
that enable the investigation of up to one million variants in cases and controls of a certain
disease (or other phenotypic traits). The HapMap was a large collaborative project that
described the frequencies of genetic variants with a minor allele frequency above 5% in four
distinct populations: Han Chinese, Japanese, Black African from Nigeria, and Caucasian of
European ancestryfrom the USA (International HapMap consortium, 2005; International
HapMap consortium, 2007).
The first GWAS to be conducted under these modalities was a large case-control study by
the Wellcome Trust Case-Control Consortium (WTCCC), in which 14,000 cases affected by 7
among the most common complex diseases (CAD, arterial hypertension, rheumatoid
arthritis, Crohn’s disease, bipolar disorder, and diabetes mellitus types I and II) were
compared with a set of 3,000 healthy controls. (Wellcome trust case control consortium,
4 Advances in the Diagnosis of Coronary Atherosclerosis

2007) The WTCCC study identified 24 genetic variants associated with at least one of these
complex diseases and helped to clarify key methodological issues, setting the stage for the
more than 400 GWAS that were to follow. These GWAS have so far identified more than 250
loci at which common variants influence the predisposition to diseases that are common
(i.e., diabetes, autoimmune diseases, and several types of cancer), an achievement that by far
outweighed that of the previous decade of genetic studies. Results are available in the
catalogue of published GWAS prepared by the National Cancer Institute (NCI)-National
Human Genome Research Institute (NHGRI). (Hindorff et al., 2010) The genetic variants
that can be identified by GWAS are common variants (with at least 5% frequency in the
population) and have a low effect size; the conferred relative risks, as expressed by odds
ratio, usually range between 1.1 and 1.5. These results confirm the views that the genetic
predisposition to common diseases consists of the combined effect of numerous common
genetic variants, each of a small effect size. However, it should be noted that GWAS identify
regions of the genome (loci) rather than variants of specific genes. Indeed, the specific
variant(s) identified by GWAS may simply represent the signal of one or more hidden
variant(s) (not typed in the arrays used in GWAS). Limitations of GWAS need to be
mentioned. First, these studies need very large samples of cases and controls. Second, DNA
and data quality control procedures and statistical analysis need to be carried out by expert
centers. Third, the overall cost of GWAS, ranging from hundreds of thousands to millions of
US dollars, is prohibitive for most research groups worldwide. And finally, even after and in
spite of all quality control procedures, there is still the chance that the results of GWAS
include false-positive results, so that an independent replication of these results is still
important even after testing thousands of individuals (Pier et al., 2010)

3. Candidate genetic factors


CAD is a complex, multifactorial disorder in which interactions among various genetic and
environmental influences play an important role. Many genes are likely to be involved in
some way in the process for CAD (Table 1). Some genetic studies have suggested that
several gene polymorphisms, including those in the genes for angiotensin-converting
enzyme (Samadi et al., 2009) and paraoxonase (Fallah et al., 2010) increase the risk for CAD.
Polymorphisms in the genes for insulin-like growth factor-I and lipoprotein lipase have
been shown to increase the risk of both CAD and type 2 diabetes (Wang et al., 1996;
Vaessen et al., 2001).
There are certain genetic defects that affect activities of some enzymes (cystathionin β-
synthase, methyltransferase and 5, 10-methylenetetrahydrofolate reductase) which may lead
to homocystinuria. Heterozygosity for deficiency of cystathionin β-synthase is known to be
linked with arthrosclerosis and thrombotic disease including CAD (Bakir et al., 2001).
Adiponectin gene locus, chromosome 3q27, is the candidate site for CAD. Adiponectin
I164T mutation is associated with the metabolic syndrome and coronary artery disease. The
I164T mutation in the adiponectin gene is reported to be a common genetic background
associated with the metabolic syndrome and CAD in the Japanese population (Ohashi et al.,
2004)
The Von Willebrand factor (VWF) may be causally associated with coronary heart disease or
merely be a marker of endothelial damage. The G allele of the -1793 C/G promoter
polymorphism in the VWF gene has been associated with higher plasma levels of VWF. Van
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 5

der Meer et al., (Van der Meer et al., 2004) found a clear association of G allele of the -1793
C/G polymorphism in the VWF gene with an increased risk of Coronary heart disease.

Location Gene name/Polymorphisms


17q23 Angiotensin-Converting Enzyme
insertion/deletion (intron 16)
1q42-q43 Angiotensinogen Met235Thr, −6G/A
3q21-q25 Angiotensin II type1 Receptor 1166A/C
8q21-q22 Aldosterone Synthase (CYP11B2) −344T/C, Lys173Ar
14q32.1-q32.2 Bradykinin B2 receptor gene −58T/C
6p24.1 Endothelin-1 Lys198Asn
7q36 eNOS Glu298Asp, −786T/C
17q21.32 Glycoprotein IIIa P1A1/A2
5q23-31 Glycoprotein Ia 807T/C
17pter-p12 Glycoprotein Ibα Thr145Met
4q28 βfibrinogen −455G/A
11p11-q12 Prothrombin 20210G/A
7q21.3-q22 PAI-1 4G/5G (promoter region)
7q21.3 Paraoxonase1 Arg192Gln, Leu54Met
8p12-p11.2 Werner Helicase Gene Cys1367Arg
1p36.3 Methylenetetrahydrofolate reductase 677C/T
16q24 NADH/NADPH oxidase p22phox242C/T, 640A/G
5q31.1 CD14 Monocyte Receptor −260C/T
11q22.3 Stromelysin (MMP3) 5A/6A (promoter region)
20q11.2-q13.1 Gelatinase B (MMP9) −1562C/T
19q13.2 ApolipoproteinE E2/E3/E4
16q21 Cholesteryl Ester Transfer Protein (CETP) Ile405Val
9q31.1 ABCA1 gene Ile823Met
3p25 PPAR-gamma Pro12Ala, Pro115Gln
20q13.11-q13.13 Prostacyclin synthase gene
Table 1. The common genetic polymorphisms which are thought to be associated with
myocardial infarction or coronary artery disease.

4. Novel genetic risk factors


4.1 MLXIPL
Triglycerides are produced from either fatty acids obtained directly from the diet or
synthesized de novo when excess carbohydrates are consumed. Genes that respond to
glucose contain a specific regulatory site, the carbohydrate response element (ChoRE), in
their promoter regions. To date, ChoREs have been mapped within the promoter regions of
the liver-type pyruvate kinase (PK), S14, fatty acid synthase (FAS), acetyl-CoA carboxylase 1
(ACC), and thioredoxin-interacting protein genes (Towle et al., 2005; Minn et al., 2005).
ChREBP is a basic helix-loop helix/leucine zipper transcription factor involved in mediating
glucose-responsive gene activation (Yamashita et al., 2001). Mice with a disruption of the
6 Advances in the Diagnosis of Coronary Atherosclerosis

ChREBP gene or hepatocytes treated with siRNA to reduce ChREBP expression cannot
induce lipogenic gene expression in response to carbohydrate (Iizuka et al., 2004; Dentin et
al., 2004). In hepatocytes prepared from ChREBP null mice, the induction can be restored by
the addition of a ChREBP expression vector (Ishii et al., 2004). Thus, ChREBP is essential for
regulating lipogenic gene expression. However, it has previously reported that ChREBP
requires an interaction partner, Mlx, to efficiently bind to ChoRE sequences and exert its
functional activity (Stoeckman et al., 2004). Mlx is a basic helix-loop helix/leucine zipper
protein that heterodimerizes with several partners, including ChREBP; MondoA, a paralog
of ChREBP expressed predominantly in skeletal muscle; and the repressors Mad1, Mad4,
and Mnt (Billin et al., 2000; Billin et al., 1999; Meroni et al., 2000). Expressing a dominant
negative form of Mlx in hepatocytes completely inhibits the glucose response of a number of
lipogenic enzyme genes, including PK, S14, ACC, and FAS (Ma et al., 2005). This inhibition
is rescued by overexpressing ChREBP but not MondoA. Therefore, Mlx is an obligatory
partner of ChREBP in regulating glucose-responsive lipogenic enzyme genes.
Recently, SNPs localized within the MLXIPL (MLX intracting protein like; ChREBP,
carbohydrate response element binding protein) loci have been associated with plasma
triglycerides (Kooner et al., 2008; Kathiresan et al., 2008). The most significant association
was described for the rs3812316 SNP (C771G, His241Gln); the CC genotype was associated
with elevated TGs. The identified SNP is located at evolutionary conserved domain
responsible for glucose dependent activation of MLXIPL. After activation and binding to the
MLX, the complex increases the transcription of genes involved, among others, in
lipogenesis and triglyceride synthesis. Since, elevated plasma triglycerides (TG) are an
independent risk factor for cardiovascular disease development (Sarwar et al., 2007) and
MLXIPL loci have been associated with plasma triglycerides, this gene might be a novel
genetic risk factor for coronary artery disease ( Pan et al., 2009)

4.2 Resistin
Resistin is a 10 kDa protein composed of 94 amino acids. It was cloned in 2001 and was
shown to be a thiazolidinedione (TZD)-regulated cytokine expressed in adipose tissue (Wolf
et al., 2004). The effect of resistin on insulin action has been extensively investigated in
laboratory models. It was shown to be involved in hepatic glucose and lipid metabolism and
appears to play a pivotal role in hepatic insulin resistance (IR) induced by high-fat diet
(Rajala et al., 2003). Resistin was suggested to affect endothelial function and the migration
of vascular smooth muscle cells (Cohen & Horel., 2009), which are regarded as key
pathophysiological mechanisms of atherosclerosis. Further, resistin has been noted to play a
vital role in increasing the level of very low density lipoprotein (VLDL) and low density
lipoprotein (LDL) in an obese person (Rizkalla et al., 2009) which is directly atherogenic.
Resistin induces increases in MCP-1 and sVCAM-1 expression in vascular endothelial cells
which suggest a possible mechanism that contribute to atherogenesis (Cohen & Horel.,
2009). Recent reports indicate that resistin promotes proliferation of VSMC that occurs
through both ERK 1/2 and Akt signalling pathways (Calabro et al., 2004). Thus resistin is
noted to enhance VSMC migration, which is a known component of athermanous plaque
synthesis (Verma et al., 2003). Resistin promotes foam cell formation via dysregulation of
scavenger receptors (SR-A) and ATP-binding cassette transporter-A1 (ABCA1) (Lee et al.,
2009) through PPAR gamma. In atherosclerosis, increased level of resistin causes elevation
of soluble TNF-_ receptor 2, IL-6 and lipoprotein-associated phospholipase A2 (Lp- PLA2)
(Reilly et al., 2005).
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 7

With respect to the reported resistin variants, the mostly extensively studied has been the
promoter variant SNP-420C>G. Functional binding studies have been done with stimulatory
proteins (Sp)-1 and 3, which bind to the promoter. Their binding has been described to be
influenced by SNP-420C>G. Sp-1 and 3 were discovered to bind efficiently only to the G-
allele sequence and after binding to increase the activity of the promoter. (Chung et al.,
2005). It seems likely that the more active promoter with the SNP-420C>G G allele is the
reason for several observations of higher plasma resistin concentration in the G allele
carriers (Yamauchi et al., 2008). However, in contrast to these studies, it has also been
reported that the genotypes of SNP-420C>G do not influence the plasma resistin
concentration in Italian subjects (Norata et al., 2007). Furthermore in a small study of
polycystic ovary syndrome patients noassociation was detected between SNP-420C>G
genotype and the serum level of resistin (Escobar-Morreale et al., 2006).
Recent studies have shown that the resistin levels are significantly correlated with coronary
artery calcification and are predictive of coronary atherosclerosis in humans (Mohty et al.,
2009). Previous studies described the association among this -420 (C>G) polymorphism, the
resistin levels and cardiovascular risk factors (Ukkola et al., 2006; Norata et al., 2007)
However, the association between the serum resistin levels and CHD seemed to be negative,
and might be controversial for this polymorphism and CAD. (Norata et al., 2007; Kunnari et
al. 2005) Differences in the cohorts might explain the different results, depending on which
ethnic group was tested (Menzaghi et al., 2006; Hivert et al., 2009). Indeed, methodological
limitations in the commercially available ELISA assays might also result in variations among
serum levels, which might cause difficulties when comparing results from different
publications.

4.3 Renalase
The kidney, in addition to maintaining fluid and electrolyte homeostasis, performs essential
endocrine functions (Peart et al., 1977). Patients with end-stage renal disease are at high risk
for cardiovascular events, even when provided optimal renal replacement therapy (Go et al.,
2004; Anavekar et al., 2004). It has been suggested that failure to replicate the endocrine
functions of the kidney may contribute to this risk, in association with heightened
sympathetic tone (Joles & Koomans 2004; Neumann et al., 2004; Wolfe et al., 1999). Renalase,
a flavin adenine dinucleotide-dependent amine oxidase that is secreted into the blood by the
kidney, metabolizes circulating catecholamines, and is deficient in chronic kidney disease
(Xu et al., 2005). Excess catecholamines promote the activity, secretion, and synthesis of
renalase, providing a novel pathway of negative feedback homeostatic control (Li et al.,
2008). In rodents, parenteral administration of renalase lowers blood pressure, heart rate,
and cardiac contractility (Xu J, Desir GV 2007). During cardiac ischemia in rats, infusion of
recombinant renalase reduces myocardial infarct size whereas neonatal nephrectomy leads
to elevated sympathetic nervous system activity, renalase deficiency, and cardiac
hypertrophy (Desir 2008; Ghosh et al., 2008). Human renalase is encoded by a 311Kbp gene
with 10 exons located on chromosome 10q23.33. The major isoform of renalase contains 342
amino acids comprising a signal peptide (amino acids 1–17), a flavin-adenine dinucleotide
(FAD) binding domain (amino acids 4–45), and a monoamine oxidase domain (amino acids
75–342). Evidence exists for at least four alternatively- spliced isoforms of renalase (Desir,
2009). The most common isoform (renalase1) is encoded by exons 1–4, 6–7, and 9. It is the
predominant human renalase protein detectable in plasma, kidney, heart, skeletal muscle,
8 Advances in the Diagnosis of Coronary Atherosclerosis

and liver. The functional significance of the spliced isoforms is not known. It has weak AA
similarities to MAO-A and MAO-B and distinct substrate specificity and inhibitor profile,
which indicates that it represents a new class of FAD-containing monoamine oxidases.
MAO-A and MAO-B are FAD containing, mitochondrial enzymes that metabolize
intracellular catecholamines. MAO-A and MAO-B have overlapping substrate specificity;
catabolize neurotransmitters such as epinephrine, norepinephrine, serotonin, and
dopamine; and are specifically inhibited by clorgyline and deprenyl, respectively.
Polyamine oxidase, the other known FAD-containing oxidase, is an intracellular oxidase
that metabolizes spermine and spermidine and regulates cell growth (Jalkanen & Salmi,
2001). Unlike MAO-A and MAO-B, which are anchored through the carboxyl terminus to
the outer mitochondrial membrane (Binda et al., 2002) and confined to intracellular
compartments, renalase is secreted into the blood, where it is detectable by Western blotting.
Amine oxidase activity has been measured in human plasma and is believed to be mediated
by vascular adhesion protein 1 (VAP-1), a copper-containing semicarbazide-sensitive amine
oxidase that is secreted by smooth muscle cells, adipocytes, and endothelial cells (Salmi &
Jalkanen, 2001). VAP-1’s substrate specificity and inhibitor profile are very different from
that of renalase. It metabolizes benzylamine and methylamine and is inhibited by
semicarbazide and hydroxylamine. Therefore, renalase is the only known amine oxidase
that is secreted into blood and that metabolizes circulating catecholamines. While the
hypotensive effect of renalase can be fully accounted for by the observed decrease in
contractility and heart rate, we cannot categorically exclude the possibility that renalase’s
effect may be partly receptor mediated.
A common missense polymorphism in the flavin-adenine dinucleotide-binding domain of
human renalase (Glu37Asp) has recently been described. This is the only reported common
coding single-nucleotide polymorphism in the renalase gene, and was recently found to be
associated with essential hypertension (Zhao et al., 2007). Whether common genetic
variation at this locus affects cardiac structure, function, and ischemia in humans is not
known.

4.4 P-selectin
P-selectin (GMP-140; granule membrane protein-140) is an adhesion molecule which
mediates the interaction of activated endothelial cells or platelets with leukocytes. The
selectin family of adhesion molecules also comprises E- and L-selectin. The genes coding for
the three selectins are clustered on chromosome 1q21–q24 (Watson et al., 1990). The P-
selectin gene spans >50 kb and contains 17 exons, most of which encode structurally distinct
domains. P-selectin is stored in a-granules of platelets and the Weibel–Palade bodies of
vascular endothelial cells (McEver et al., 1989) it rapidly shifts from the membranes of
secretory granules to the surface of platelets and endothelial cells upon stimulation by
oxidized low density lipoprotein (LDL) (Vora et al., 1997), oxygen radicals (Patel et al.,
1991), thrombin (Lorant et al., 1991), cytokines and various other stimuli (Zimmermann, et
al., 1990). P-selectin is required for efficient recruitment of neutrophils in acute and chronic
inflammation (Johnson, R.C., et al. 1995) and recently has been shown to bind T cells on
vascular endothelial cells. These properties suggest that P-selectin could contribute to
atherogenesis (Hansson, G.K. , 1989, Libby, P. and Hansson, G.K.,1991). Actually, P-selectin
expression has been demonstrated to be significantly increased in endothelium overlying
atherosclerotic plaques, and it is focally expressed in the aorta of hypercholesterolemic
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 9

rabbits. It has been reported that P-selectin-deficient mice on an atherogenic diet develop
significantly smaller fatty streaks than non-deficient mice (Johnson, R.C., et al. 1997). In
humans, plasma P selectin levels have been shown to be increased in diabetic patients, in
patients with unstable angina, post-angioplasty restenosis and after coronary artery spasm
(Kaikita, K. , 1995).
Recent study that genotyped 5 single nucleotide polymorphisms (SNPs) in P-selectin (SELP)
(V168M, S290N, N592D, V599L, T715P), 2 SNPs (M62I, S273F) in P-selectin glycoprotein
ligand-1 (SELPLG), 5 SNPs in CD40LG (−3459A>G, −122A>C, −123A>C, 148T>C, intr4–
13T>C), the H558R SNP in SCN5A, and rs2106261 in ZFHX3. In addition, length
polymorphisms in SELPLG (36bp-tandem repeat) and CD40LG (CA-repeat) were genotyped
by PCR methods. None of the gene polymorphisms showed significant differences between
AMI patients and healthy controls. Among patients with a history of VF (Ventricular
fibrillation), however, the SELP 168M variant showed a significantly higher prevalence as
compared with patients without VF. This was the first description of an association of the
SELP gene variant 168M with primary VF during acute MI. This variant may be a novel
polymorphism for evaluating the susceptibility for VF in the setting of acute MI. (Elmas, 2010)

4.5 KDR
Kinase insert domain-containing receptor/fetal liver kinase-1, also called VEGFR2 (KDR), is
expressed in a wide variety of cells such as endothelial progenitor cells (EPCs), endothelial
cells, and primitive and more mature hematopoietic cells. Kinase insert domain-containing
receptor/ fetal liver kinase-1 is required for the differentiation of EPCs and for the
movement of EPCs from the posterior primitive streak to the yolk sac, a precondition for the
subsequent formation of blood vessels (Shalaby et al., 1997). Studies with KDR knockout
mice have found that KDR plays critical roles in the development and formation of blood
vessel networks (Fong et al., 1995). Vascular endothelial growth factor binds to 2 tyrosine
kinase receptors, VEGF receptor-1 (VEGFR1, Flt-1) and KDR, in endothelial cells. The
mitogenic and chemotactic effects of VEGF are mediated mainly through KDR in
endothelial cells VEGF receptor signal transduction which is activated through
autophosphorylation of tyrosine residues in the cytoplasmic kinase domain of KDR. This
event is followed by activation of downstream signaling pathways such as mitogen-
activated protein kinases, Akt and eNOS, which are essential for migration and proliferation
of endothelial cells, thereby stimulating angiogenesis (Matsumoto & Claesson-Welsh, 2001).
After vascular endothelial growth factor (VEGF) binding to KDR, multiple early signaling
cascades are activated in EPCs and in endothelial cells. An array of biological activities are
subsequently elicited in vivo and in vitro, including angiogenesis, endothelial survival,
proliferation, migration, and increased production of nitric oxide and prostaglandin I2
(Gerber et al., 1994). Dysregulated vessel growth is implicated in the pathogenesis of a wide
variety of diseases, including proliferative retinopathies, tumors, rheumatoid arthritis,
atherosclerosis, as well as CHD. (Dimmeler et al., 2001; Rehman et al., 2004; Werner et al.,
2002). The variation of KDR gene may change the biological function of KDR. Bioinformatic
analysis showed that the single nucleotide polymorphism (SNP) -604T/C (rs2071559) leads
to structural alteration of the binding site for transcriptional factor E2F (involving in cell
cycle regulation, interacting with Rb p107 protein) in KDR gene promoter region, which
may alter KDR expression. Exonic polymorphisms SNP1192G/A (rs2305948, in exon 7) and
SNP1719A/T (rs1870377, in exon 11) are located in the third and fifth NH2-terminal IG-like
10 Advances in the Diagnosis of Coronary Atherosclerosis

domains within the extracellular region, which are important for ligand binding, and result
in nonsynonymous amino acid changes at residue 297V/I and 472H/Q, respectively. It is
showed that patients carrying the KDR mutations are more susceptible to CHD. The higher
CHD risk could be due to downregulation of the VEGF/KDR signaling pathway. However,
the angiogenesis preceded by VEGF/KDR signaling pathway could be decreased due to low
KDR activity. The SNP-604T/C decreases mRNA levels of KDR and both SNP1192G/A and
SNP1719T/A resulted in slight but significant decrease in the VEGF binding efficiency to
KDR. One can speculate that the decrease in KDR function is correlated with vascular
dysfunction, including endothelial cell damage, impaired endothelial cell survival,
decreased antiapoptotic effects of VEGF, and abnormal vascular repair. All of these can
promote the progression of atherosclerotic disease. A previous study has suggested that -
907T/C, -11903G/A, and -18487A/T (now called SNP-604, SNP1192, and SNP1719,
respectively) have no significant association with the development of coronary artery lesion
in Japanese subjects with Kawasaki disease (Kariyazono et al., 2004). The differences
between the studies could be due to the different pathologic mechanism between the
coronary artery lesion with Kawasaki disease and CHD, different genetic background of the
populations as well as the sample size in different studies. (Schmidt-Lucke et al., 2005). The
genotype frequencies of the 3 SNPs from the HapMap data were similar to others. The
rs2071559 can capture rs7667298 (exon_1, untranslated). No linkage disequilibrium was
found in rs2305948 with other SNPs in HapMap CHB data. The rs1870377 (exon_11) can
capture rs10016064 (intron_13), rs17085265 (intron_21), rs3816584 (intron_16), rs6838752
(intron_17), rs1870379 (intron_15), rs2219471 (intron_20), rs1870378 (intron_15), rs13136007
(intron_13), and rs17085262 (intron_21). The results showed that 2 blocks were captured by
SNP-604 (rs2071559) and SNP1719 (rs1870377), respectively, and SNP1192 (rs2305948) were
associated with CHD.

5. Other novel polymorphisms


Asymmetrical dimethylarginine (ADMA), an endogenous arginine analogue, inhibits nitric
oxide synthases and plays an important role in endothelial dysfunction. The results suggest
that the DDAH1 (dimethylarginine dimethylaminohydrolase 1) loss-of-function
polymorphism is associated with both increased risk of thrombosis stroke and CAD (Ding et
al., 2010). Growing evidence has shown that inflammation plays crucial roles in the
development of coronary artery disease. Interleukin-16 (IL-16), a multifunctional cytokine, is
involved in a series of inflammatory disorders. One finding indicates that IL-16 may be used
as a genetic marker for CAD susceptibility (Wu et al., 2010). Two single-nucleotide
polymorphisms (SNPs), rs1746048 and rs501120, from genome wide association studies of
coronary artery disease map to chromosome 10q11 ~80 kb downstream of chemokine
CXCL12. Coronary artery disease risk alleles downstream of CXCL12 are associated with
plasma protein levels of CXCL12 and appear to be related to CXCL12 transcript levels in two
human cell lines. This implicates CXCL12 as potentially causal and supports CXCL12 as a
potential therapeutic target for CAD ( Nehal et al., 2011).

6. Conclusion
Studying only one SNP may be an overly simplistic method in investigating a complex
disease such as CAD. Complex traits such as CAD, more large databases of high-quality
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 11

genetic and clinical data need to be established and since genes and environmental factors
are both involved in this disease; environmental causes and gene-environment interactions
must be carefully assessed. These results will provide clues to the involvement and
investigation of novel candidate genes in association studies. Large replication studies with
different ethic samples are needed to investigate whether there are ethnic differences in the
influence of novel polymorphisms on coronary artery disease. This issue should be
investigated with different ethnic samples in the future. Therefore studies with large sample
size of coronary artery disease and different ethnicity will be welcome to help elucidate the
interconnection between novel genetics and pathogenesis of coronary artery disease.

7. Abbreviations
ABCA1: ATP-binding cassette transporter 1
ACC: acetyl-CoA carboxylase
ALOX5AP: Arachidonate 5-lipoxygenase-activating protein.
APOA1: Apolipoprotein A1
CAD: Coronary artery disease
LCAT: Lecithin-cholesterol acyltransferase
CHD: Coronary heart disease
ChoRE: Carbohydrate response element
ChREBP: Carbohydrate response element binding protein
ELISA: Enzyme-linked immunosorbent assay
EPCs: Endothelial progenitor cells
ERK 1/2 : Extracellular signal-regulated kinases1/2
FAD: Flavin-adenine dinucleotide
FAS: Fatty acid synthase
GWAS: Genome-wide association Studies
HDL: High-density lipoprotein
KDR: Kinase insert domain-containing receptor
Lp- PLA2: Lipoprotein-associated phospholipase A2
Mad1: Mitotic arrest deficient-like 1
Mad4: Mitotic arrest deficient-like 4
MAO-A: L-Monoamine oxidases A
MAO-B : L-Monoamine oxidases B
MEF2A: Myocyte enhancer factor 2A
MlX: Max-like x protein
PPAR: Peroxisome proliferator-activated receptors
PK : Pyruvate kinase
siRNA: Small interfering RNA
SNPs: Single nucleotide polymorphisms
Sp1: stimulatory proteins 1
SR-A: Scavenger receptors A
TNF: Tumor necrosis factors
VAP-1: vascular adhesion protein 1
VCAM-1: Vascular cell adhesion protein 1
VF: Ventricular fibrillation
VLDL: Very low density lipoprotein
VWF: Von Willebrand factor
12 Advances in the Diagnosis of Coronary Atherosclerosis

8. References
Anavekar, N.S. (2004). Relation between renal dysfunction and cardiovascular outcomes
after myocardial infarction, N. Engl. J. Med. 351:1285–1295.
Bakir, I., Zarzou W and Herrmann, W. (2001). Genetic Polymorphism of 5,10-Methylene-
tetrahydrofolate Reductase (MTHFR) as Risk Factor for Coronary Artery Disease,
Arab Journal of Pharmaceutical Sciences 1: 41-50.
Berliner J.C. (1997). Induction of P-selectin by oxidized lipoproteins. Separate effects on
synthesis and surface expression, Circ. Res. 80: 810–818
Bernhard, R., Winkelmann, Joerg Hager, William, E. Kraus, Piera Merlini, Bernard Keavney,
Peter, J., Grant, Joseph B. Muhlestein, and Christopher B. Granger. (2000). Genetics
of coronary heart disease: Current knowledge and research principles, Am heart J
140: 511-526
Billin, A. N., Eilers, A. L., Queva, C., and Ayer, D. E. (1999). Mlx, a novel Max-like BHLHZip
protein that interacts with the Max network of transcription factors, J. Biol. Chem.
274: 36344–36350
Billin, A. N., Eilers, A. L., Coulter, K. L., Logan, J. S., and Ayer, D. E. (2000). Mondo A, a
Novel Basic Helix-Loop-Helix-Leucine Zipper Transcriptional Activator That
Constitutes a positive Branch of a Ma-Like Network, Mol. Cell. Biol. 20: 8845–8854
Binda, C., Mattevi, A., and Edmondson, D.E. (2002). Structure-function relationships in
flavoenzyme dependent amine oxidations: a comparison of polyamine oxidase and
monoamine oxidase, J. Biol. Chem. 277:23973–23976.
Broeckel, U., Hengstenberg, C., Mayer, B., Holmer, S., Martin, LJ., Comuzzie, AG., et al.
(2002). A comprehensive linkage analysis for myocardial infarction and its related
risk factors, Nat Genet 30: 210-4.
Calabro, P., Samudio, I., Willerson ,JT., Yeh, ET. (2004). Resistin promotes smooth muscle
cell proliferation through activation of extracellular signal regulated kinase 1/2 and
phosphatidylinositol 3-kinase pathways, Circulation 110: 3335–3340.
Chung, SS., Choi, HH., Kim, KW., Cho, YM., Lee, HK & Park KS .(2005). Regulation of
human resistin gene expression in cell systems: an important role of stimulatory
protein 1 interaction with a common promoter polymorphic site, Diabetologia 48:
1150–1158.
Cohen, JC., Kiss, RS., Pertsemlidis, A., Marcel, YL., McPherson, R., Hobbs, HH. (2004).
Multiple rare alleles contribute to low plasma levels of HDL cholesterol, Science
305 : 869-72.
Cohen, G., Hörl, WH. (2009). Resistin as a cardiovascular and atherosclerotic risk factor and
uremic toxin, Semin. Dial., 22: 373-377.
Dentin, R., Pegorier, J. P., Benhamed, F., Foufelle, F., Ferre, P., Fauveau, V., Magnuson, M.
A., Girard, J., and Postic, C. (2004). Hepatic glucokinase is required for the syner-
gistic action of ChREBP and SREBP-1c on glycolytic and lipogenic gene expression,
J. Biol. Chem. 279: 20314–20326
Desir, GV. (2008). Renalase deficiency in chronic kidney disease, and its contribution to
hypertension and cardiovascular disease, Curr Opin Nephrol Hypertens 17: 181–185.
Desir, GV. (2009). Regulation of blood pressure and cardiovascular function by renalase,
Kidney Int 76: 366–370.
Ding, H., Wu, B., Wang, H., Lu, Z., Yan, J., Wang, X., Shaffer, JR., Hui, R., Wang DWA.
(2010). Novel loss-of-function DDAH1 promoter polymorphism is associated with
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 13

increased susceptibility to thrombosis stroke and coronary heart disease, Circ Res.
106(6): 1019-21.
Escobar-Morreale, HF., Villuendas, G., Botella-Carretero, JI., Alvarez-Blasco, F., Sanchón , R.,
Luque-
Norata, GD., Ongari, M., Garlaschelli, K., et al. (2007). Effect of the -420C/G variant of the
resistin gene promoter on metabolic syndrome, obesity, myocardial infarction and
kidney dysfunction, J Intern Med 262: 104–112.
Fallah, S., Seifi, M., Ghasemi, A., Firoozrai, M., Samadikuchaksaraei, A. (2010). Matrix
Metalloproteinase-9 and Paraoxonase 1 Q/R192 Gene Polymorphisms and the Risk
of Coronary Artery Stenosis in Iranian Subjects, J. Clin. Lab. Anal. 24: 305–310,
Farrall, M., Green, FR., Peden, JF., Olsson, PG., Clarke, R., Hellenius ML., et al. (2006).
Genome-wide mapping of susceptibility to coronary artery disease identifies a
novel replicated locus on chromosome 17, PLoS Genet 2: e72.
Fong, GH., Rossant, J., Gertsenstein, M., Breitman, ML,. (1995). Role of the Flt-1 receptor
tyrosine kinase in regulating the assembly of vascular endothelium, Nature 376: 66 –
70.
Frazer KA, Ballinger DG, Cox DR et al. (2007). A second generation human haplotype map
of over 3.1 million SNPs, Nature 449: 851-861.
Frikke-Schmidt, R., Nordestgaard, BG., Jensen, GB., Tybjaerg-Hansen, A. (2004). Genetic
variation in ABC transporter A1 contributes to HDL cholesterol in the general
population, J Clin Invest 114 : 1343-53.
Gerber, HP., McMurtrey, A., Kowalski, J., et al. (1998). Vascular endothelial growth factor
regulates endothelial cell survival through the phosphatidylinositol 3=-kinase/Akt
signal transduction pathway: requirement for Flk-1/KDR activation, J Biol Chem
273: 30336–43.
Ghosh, SS., Krieg, RJ., Sica, DA., Wang, R., Fakhry, I., et al. (2009). Cardiac hypertrophy in
neonatal nephrectomized rats: the role of the sympathetic nervous system. Pediatr
Nephrol. 24: 367–377.
Go, A.S., Chertow, G.M., Fan, D., McCulloch, C.E., and Hsu, C.Y. (2004). Chronic kidney
disease and the risks of death, cardiovascular events, and hospitalization, N. Engl. J.
Med. 351: 1296–1305.
Hansson, G.K., Jonasson, L., Seifert, P.S. and Stemme, S. (1989) Immune mechanisms in
atherosclerosis. Arteriosclerosis 9: 567–578.
Hauser, ER., Crossman, DC., Granger, CB., Haines, JL., Jones, C.J, Mooser, V., et al. (2004). A
genomewide scan for early-onset coronary artery disease in 438 families: the
GENECARD Study, Am J Hum Genet 75: 436-47.
He, J., Gu D, Wu X, Reynolds K, Duan X, Yao C, Wang J, Chen CS, Chen J, Wildman RP et al.
(2005). Major causes of death among men and women in China, N Engl J Med 353:
1124-1134.
Helgadottir, A., Manolescu, A., Thorleifsson, G., Gretarsdottir, S., Jonsdottir, H.,
Thorsteinsdottir, U., et al. (2004). The gene encoding 5-lipoxygenase activating
protein confers risk of myocardial infarction and stroke, Nat Genet 36: 233-9.
Helgadottir, A., Gretarsdottir, S., St Clair, D., Manolescu, A., Cheung, J., Thorleifsson, G., et
al. (2005). Association between the gene encoding 5-lipoxygenase-activating protein
and stroke replicated in a Scottish population, Am J Hum Genet 76 : 505-9.
14 Advances in the Diagnosis of Coronary Atherosclerosis

Hill, JM., Zalos, G., Halcox, JP., et al. (2003). Circulating endothelial progenitor cells,
vascular function, and cardiovascular risk, N Engl J Med 348 :593– 600.
Hindorff, LA., Junkins, HA., Mehta, JP., Manolio, TA. (2010). A catalog of published
genome-wide association studies. www.genome.gov/ gwastudies
Hivert, MF., Manning, AK., McAteer, JB., et al. (2009). Association of variants in RETN with
plasma resistin levels and diabetes-related traits in the Framingham Offspring
Study, Diabetes 58: 750–756.
Iizuka, K., Bruick, R. K., Liang, G., Horton, J. D., and Uyeda, K. (2004). Deficiency of
carbohydrate response element binding protein (ChREBP) reduces lipogenesis as
well as glycolysisProc, Natl. Acad. Sci. U. S. A. 101: 7281–7286
International HapMap consortium. (2005). A haplotype map of the human genome, Nature
437: 1299-1320.
International HapMap comsortium. (2007). A second generation human haplotype map of
over 3.1 million SNPs, Nature 449: 851-861.
Ishii, S., Iizuka, K., Miller, B. C., and Uyeda, K. (2004). Carbohydrate response element
binding protein directly promotes lipogenic enzyme gene transcription, Proc. Natl.
Acad. Sci. U. S. A. 101: 15597–15602
Jalkanen, S., and Salmi, M. (2001). Cell surface monoamine oxidases: enzymes in search of a
function, EMBO J. 20:3893–3901.
JC., and Gomez Garcia, EB. (2004). Genetic variability of von Willebrand factor and risk of
coronary heart disease: the Rotterdam Study, Br J Haematol 124: 343-347.
Jian, Wu., Yin, Wang., Yi, Zhang., Longjiang, Li. (2010). Association Between IL-16
Polymorphisms and Risk of Coronary Artery Disease, DNA and Cell Biology . 00. (0)
1-1.
Johnson, R.C., Mayadas, T.N., Frenette, P.S., Mebius, R.E., Subramaniam, M., Lacasce, A.,
Hynes, R.O. and Wagner, D.D. (1995) Blood cell dynamics in P-selectin-deficient
mice, Blood 86: 1106–1114.
Johnson, R.C., Chapman, S.M., Dong, Z.M., Ordovas, J.M., Mayadas, T.N., Herz, J., Hynes,
R.O., Schaefer, E.J. and Wagner, D.D. (1997). Absence of P-selectin delays fatty
streak formation in mice, J. Clin. Invest. 99: 1037–1043.
Joles, JA., Koomans, HA. (2004). Causes and consequences of increased sympathetic activity
in renal disease, Hypertension 43: 699–706.
Kaikita, K., Ogawa, H., Yasue, H., Sakamoto, T., Suefuji, H., Sumida, H. and Okumura, K.
(1995). Soluble P-selectin is released into the coronary circulation after coronary
spasm, Circulation 92: 1726–1730.
Kariyazono, H., Ohno, T., Khajoee, V., et al. (2004). Association of vascular endothelial
growth factor (VEGF) and VEGF receptor gene polymorphisms with coronary
artery lesions of Kawasaki disease, Pediatr Res 56:953–9.
Kathiresan, S., Melander, O., Guiducci, C., Surti, A., Burtt, NP., Rieder, MJ., et al. (2008). Six
new loci associated with blood low-density lipoprotein cholesterol, high-density
lipoprotein cholesterol or triglycerides in humans, Nat Genet 40:189–197
Kooner, JS., Chambers, JC., Aguilar-Salinas, CA., Hinds, DA., Hyde, CL., Warnes, GR., et al.
(2008). Genome-wide scan identifies variation in MLXIPL associated with plasma
triglycerides, Nat Genet 40:149–151
Kunnari, A., Ukkola, O., Kesaniemi, YA. (2005). Resistin polymorphisms are associated with
cerebrovascular disease in Finnish Type 2 diabetic patients, Diabet Med 22: 583–589.
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 15

Lander, ES. (1996). The new genomics: global views of biology, Science 274: 536-9.
Lanktree M. (2008). Genetic Basis of Coronary Artery Disease, UWOMJ 77(2): 55-58
Lee, TS., Lin, CY., Tsai, JY., Wu, YL., Su, KH., Lu, KY., Hsiao, SH., Pan, CC., Kou, YR., Hsu,
YP., Ho, LT. (2009). Resistin increases lipid accumulationby affecting class A
scavenger receptor, CD36 and ATP-binding cassette transporter-A1 in
macrophages, Life. Sci 16: 97-104.
Li, G., Xu, J., Wang, P., Velazquez, H., Li, Y., et al. (2008). Catecholamines regulate the
activity, secretion, and synthesis of renalase, Circulation 117: 1277–1282.
Libby, P. and Hansson, G.K. (1991). Involvement of the immune system in human
atherosclerosis: current knowledge and unanswered questions, Lab. Invest. 64: 5–15.
Lusis, AJ., Mar, R., Pajukanta P. (2004). Genetics of atherosclerosis, Annu Rev Genomics Hum
Genet 5: 189-218.
Ma, L., Tsatsos, N. G., and Towle, H. C. (2005). Direct Role of ChREBP/Mlx in Regulating
Hepatic Glucose-responsive Genes, J. Biol. Chem. 280: 12019–12027
Mannuccio, MF., Lotta, LA., Peyvandi, F. (2010). Genome-Wide Association Studies in
Myocardial Infarction and Coronary Artery Disease, J Teh Univ Heart Ctr 3 116-121
Matsumoto, T., Claesson-Welsh, L. (2001). VEGF receptor signal transduction. Sci STKE
112:RE21.
The International HapMap Consortium. (2003). The International HapMap Project. Nature
426: 789 –96.
McEver, R.P., Beckstead, J.H., Moore, K.L., Marshall-Carlson, L. and Bainton, D.F. (1989).
GMP-140, a platelet alpha-granule membrane protein, is also synthesized by
vascular endothelial cells and is localized in Weibel–Palade bodies, J. Clin. Invest.
84: 92–99.
Menzaghi, C., Coco, A., Salvemini, L., et al. (2006). Heritability of serum resistin and its
genetic correlation with insulin resistance-related features in nondiabetic
Caucasians, J Clin Endocrinol Metab 91: 2792–2795.
Meroni, G., Cairo, S., Merla, G., Messali, S., Brent, R., Ballabio, A., and Reymond, A. (2000).
Mlx, a new Max-like bHLHZip family member: the center stage of a novel
transcription factors regulatory pathway? Oncogene 19: 3266-277
Minn, A. H., Hafele, C., and Shalev, A. (2005). Thioredoxin-Interacting Protein Is Stimulated by
Glucose through a Carbohydrate Response Element and Induces ß-Cell Apoptosis,
Endocrinology 146: 2397–2405
Mohty, D., Pibarot, P., Despres, JP., et al. (2009). Age-related differences in the pathogenesis
of calcific aortic stenosis: The potential role of resistin, Int J Cardiol in press.
Nehal, N,. Mehta, Mingyao, Li., Dilusha William, Amit V. Khera., Stephanie
DerOhannessian., Liming Qu et al. (2011). The novel atherosclerosis locus at 10q11
regulates plasma CXCL12 levels , Eur Heart J.00. (0) 1-1
Neumann, J., Ligtenberg, G., Klein, II., Koomans, HA., Blankestijn, PJ. (2004). Sympathetic
hyperactivity in chronic kidney disease: pathogenesis, clinical relevance, and
treatment, Kidney Int 65: 1568–1576.
Norata, GD., Ongari, M., Garlaschelli, K., Tibolla, G., Grigore, L., Raselli, S., et al. (2007).
Effect of the -420C/G variant of the resistin gene promoter on metabolic syndrome,
obesity, myocardial infarction and kidney dysfunction, J Intern Med 262: 104–112.
16 Advances in the Diagnosis of Coronary Atherosclerosis

Ohashi, K., Ouchi, N., Kihara, S., Funahashi, T., Nakamura, T., Sumitsuji, S., et al. (2004).
Adiponectin I164T mutation is associated with the metabolic syndrome and
coronary artery disease, J Am Coll Cardiol 43: 1195-1200.
Pajukanta, P., Cargill, M., Viitanen, L., Nuotio, I., Kareinen, A., Perola, M., et al. (2000). Two
loci on chromosomes 2 and X for premature coronary heart disease identified in
early- and late-settlement populations of Finland, Am J Hum Genet 67 : 1481-93.
Pan, LA., Chen, YC., Huang, H., Zhang, L., Liu, R., Li, X., Qiang, O., Zeng, Z,. (2009). G771C
Polymorphism in the MLXIPL Gene Is Associated with a Risk of Coronary Artery
Disease in the Chinese: A Case-Control Study, Cardiology. 114(3): 174-8.
Peart, WS. (1977). The kidney as an endocrine organ. Lancet 2: 543–548.of death,
cardiovascular events, and hospitalization, N Engl J Med 351: 1296–1305.
Pritchard, JK. (2001). Are rare variants responsible for susceptibility to complex diseases?
Am J Hum Genet 69 : 124-37.
Rajala, MW., Obici, S., Scherer, PE., Rossetti, L. (2003). Adipose-derived resistin and gut-
derived resistinlike molecule-beta selectively impair insulin action on glucose
production, J Clin Invest 111: 225–230.
Ramírez, M. & San Millán, JL. (2006). Adiponectin and resistin in PCOS: aclinical,
biochemical and molecular genetic study, Hum Reprod 21: 2257–2265.
Rehman, J., Li, J., Parvathaneni, L., et al. (2004). Exercise acutely increases circulating
endothelial progenitor cells and monocyte-/macrophagederived angiogenic cells, J
Am Coll Cardiol 43: 2314–8
Reich, DE. , Lander, ES. (2001). On the allelic spectrum of human disease, Trends Genet 17 :
502-10
Reilly, MP., Lehrke, M., Wolfe, ML., Rohatgi, A., Lazar, MA., Rader, DJ. (2005). Resistin Is an
Inflammatory Marker of Atherosclerosis in Humans, Circulation 111: 932-939.
Rizkalla, J., Melone, M., Zhao, A., Rashid, S. (2009). The Pathophysiological Role of resistin
in Impaired Lipoprotein Metabolism in Obesity, Circulation 120: S529.
Samadi Rad, B., Seifi, M., Ghasemi, A., Khosravi, M., Afghan, MR. , Daghigh Azar, B.
(2009). The association of ACE gene insertion/deletion polymorphism with
coronary artery disease in Iranian subjects, 8th International Congress on Coronary
Artery Disease – ICCAD. Czech Republic, Prague, ( Octobr, 2009): 521-527
Samani, NJ., Burton, P., Mangino, M., Ball, SG., Balmforth, AJ., Barrett J., et al. (2005). A
genomewide linkage study of 1,933 families affected by premature coronary artery
disease: The British Heart Foundation (BHF) Family Heart Study, Am J Hum Genet
77 : 1011-20.
Sarwar, N., Danesh, J., Eiriksdottir, G., Sigurdsson, G., Wareham, N., Bingham S, et al.
(2007). Triglycerides and the risk of coronary heart disease: 10 158 incident cases
among 262 525 participants in 29 Western prospective studies, Circulation 115: 450–
458
Salmi, M., and Jalkanen, S. (2001). VAP-1: an adhesion and an enzyme, Trends Immunol. 22:
211–216.
Shalaby, F., Rossant, J., Yamaguchi, TP., et al. (1995). Failure of blood-island formation and
vasculogenesis in Flk-1-deficient mice, Nature 376: 62– 6.
Shalaby, F., Ho, J., Stanford, WL., et al. (1997). A requirement for Flk1 in primitive and
definitive hematopoiesis and vasculogenesis, Cell 89: 981–90.
Mechanisms of Disease: Novel Polymorphisms in Coronary Artery Disease 17

Stoeckman, A. K., Ma, L., and Towle, H. C. (2004). Mlx is the functional heteromeri partner
of the carbohydrate response element-binding protein in glucose regulation of
lipogenic enzyme genes, J. Biol. Chem. 279 (15) : 15662–15669.
Strehlow, K., Werner, N., Berweiler, J., et al. (2003). Estrogen increases bone marrow-derived
endothelial progenitor cell production and diminishes neointima formation,
Circulation 107:3059–65.
Towle, H. C. (2005). Glucose as a Regulator of Eukaryotic Gene Transcription, Trends
Endocrinol. Metab. 16: 489–494
Takahashi, T., Yamaguchi, S., Chida, K., Shibuya, M. (2001). A single autophosphorylation
site on KDR/Flk-1 is essential for VEGF-A-dependent activation of PLC-gamma
and DNA synthesis in vascular endothelial cells, EMBO J 20: 2768 –78.
Ukkola, O., Kunnari, A., Kesaniemi, YA,. (2008). Genetic variants at the resistin locus are
associated with the plasma resistin concentration and cardiovascular risk factors,
Regul Pept 3: 56–59.
Vaessen, N., Heutink, P., Janssen, JA., Witteman, JCM., Testers, L., Hofman, A., Lamberts,
SWJ., Oostra, BA., Pols, HAP., and Van Duijn CM. (2001). A polymorphism in the
gene for IGF-I. Functional properties and risk for type 2 diabetes and myocardial
infarction, Diabetes 637-642.
Van der Meer, IM., Brouwers, GJ., Bulk, S., Leebeek, FW., van der Kuip ,DA., Hofman, A.,
Witteman, JC., Gómez García EB. (2004). Genetic variability of von Willebrand
factor and risk of coronary heart disease: the Rotterdam Study, Br J Haematol
124(3): 343-7.
Verma, S., Li, SH., Wang, CH., Fedak, PWM., Li, RK., Weisel, RD., Mickle, AG. (2003).
Resistin promotes endothelial cell activation: further evidence of adipokine
endothelial interaction, Circulation 108: 736–740
Vora, D.K, Fang, Z.-T., Liva, S.M., Tyner, T.R., Parhami, F., Watson, A.D., Drake, T.A.,
Territo, M.C. and and Berliner JA. (1997). Induction of P-Selectin by oxidized
lipoproteins-separate effects on synthesis and surface Expression Circ Res 80: 810-
818
Waltenberger, J., Claesson-Welsh, L., Siegbahn, A., Shibuya, M., Heldin, CH. (1994).
Different signal transduction properties of KDR and Flt1, two receptors for vascular
endothelial growth factor, J Biol Chem 269:26988 –95.
Wang, XL., McCredie, RM. and Wilcken DEL. (1996). Common DNA polymorphisms at the
lipoprotein lipase gene. Association with severity of coronary artery disease and
diabetes, Circulation 93:1339-1345.
Wang, L., Fan, C., Topol, SE., Topol, EJ., Wang, Q. (2003). Mutation of MEF2A in an
inherited disorder with features of coronary artery disease. Science 302: 1578-81.
Watson, M.L., Kingsmore, S.F., Johnston, G.I., Siegelmann, M.H., Le Beau M.M., Lemons,
R.S., Bora, N.S., Howard, T.A., Weissman, I.L., McEver, R.P. and Seldin, M.P. (1990)
Genomic organization of the selectin family of leukocyte adhesion molecules on
human and mouse chromsome 1, J. Exp. Med. 172: 263–272.
Wellcome trust case control consortium. (2007). Genome-wide association study of 14,000
cases of seven common diseases and 3,000 shared controls, Nature 447: 661-678.
Werner, N., Priller, J., Laufs, U., et al. (2002). Bone marrow-derived progenitor cells
modulate vascular reendothelialization and neointimal formation: effect of 3-
18 Advances in the Diagnosis of Coronary Atherosclerosis

hydroxy-3-methylglutaryl coenzyme A reductase inhibition, Arterioscler Thromb


Vasc Biol 22: 1567–72.
Werner, N., Kosiol, S., Schiegl, T., et al. (2005). Circulating endothelial progenitor cells and
cardiovascular outcomes, N Engl J Med 353: 999–1007.
Wolfe, RA., Ashby, VB., Milford, EL., Ojo, AO., Ettenger, RE., et al. (1999). Comparison of
mortality in all patients on dialysis, patients on dialysis awaiting transplantation,
and recipients of a first cadaveric transplant, N Engl J Med 341: 1725–1730.
Wolf G. (2004). Insulin resistance and obesity: Resistin, a hormone secreted by adipose
tissue, Nutr Rev 62(10): 389–394.
Wright, AF. Hastie, ND. (2001). Complex genetic diseases: controversy over the Croesus
code, Genome Biol 2 : 1-8.
Xu, J., Li, G., Wang, P., Velazquez, H., Yao, X., et al. (2005). Renalase is a novel, soluble
monoamine oxidase that regulates cardiac function and blood pressure, J Clin Invest
115: 1275–1280.
Xu, J., Desir, GV. (2007). Renalase, a new renal hormone: its role in health and disease, Curr
Opin Nephrol Hypertens 16: 373–378.
Yamashita, H., Takenoshita, M., Sakurai, M., Bruick, R. K., Henzel, W. J., Shillinglaw, W.,
Arnot, D., and Uyeda K. (2001A) glucose-responsive transcription factor that
regulates carbohydrate metabolism in the live, Proc Natl Acad Sci U S A. 98(16):
9116-21
Yamauchi, J., Osawa, H., Takasuka, T., Ochi, M., Murakami, A., Nishida, W., et al. (2008).
Serum resistin is reduced by glucose and meal loading in healthy human subjects,
Metabolism 57: 149–156.
Zdravkovic, S., Wienke, A., Pedersen, NL., Marenberg, ME., Yashin, AI and De Faire, U,.
(2002). Heritability of death from coronary heart disease: A 36-year follow-up of
20,966 Swedish twins, J Intern Med 252: 247-254.
Zhao, Q., Fan, Z., He, J., Chen, S., Li, H., et al. (2007). Renalase gene is a novel susceptibility
gene for essential hypertension: a two-stage association study in northern Han
Chinese population, J Mol Med 85: 877–885.
2

Multifunctional Role of TRAIL


in Atherosclerosis and
Cardiovascular Disease
Katsuhito Mori, Masanori Emoto and Masaaki Inaba
Department of Metabolism, Endocrinology and
Molecular Medicine Osaka City University
Graduate School of Medicine, Osaka
Japan

1. Introduction
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) belongs to the TNF
ligand superfamily. As the name suggests, TRAIL was identified and cloned based on its
sequence homology to the extracellular domain of TNF ligand family members (Pitti et al.,
1996; Wiley et al., 1995). Human TRAIL consists of 281 amino acids. TRAIL is a type II
transmembrane protein with a short intracellular amino-terminal domain and an extracellular
carboxy-terminal domain (Fig. 1). TRAIL is cleaved by cysteine proteases from the cell surface
to form a soluble ligand, and when released soluble TRAIL is measurable in human blood.
TRAIL contains cysteine residue (Cys 230) that interacts with the zinc ion, resulting in the
formation of a TRAIL homotrimer (Hymowitz et al., 1999) (Fig. 1). This trimerization is
necessary for TRAIL to bind to its receptors and to exert optimal biological activity, as is
described below. (Hao et al., 2004; Johnstone et al., 2008; Wang, 2008; Wu et al., 2004).
TRAIL binds to five different receptors found on a variety of cells. Of these receptors, two,
TRAIL-R1 (DR4) and TRAIL-R2 (DR5), contain a cytoplasmic death domain and trigger
TRAIL-induced apoptosis. Two decoy receptors lacking a functional death domain, TRAIL-
R3 (DcR1) and TRAIL-R4 (DcR2), compete with TRAIL-R1 and TRAIL-R2 for TRAIL
binding, possibly antagonizing apoptotic signaling. In addition, osteoprotegerin (OPG) is a
fifth soluble decoy receptor. Briefly, the TRAIL apoptosis pathway is initiated by the
binding of a TRAIL trimer to TRAIL-R1 or TRAIL-R2, which leads to receptor trimerization.
This receptor conformational change recruits the adaptor protein Fas-associated death
domain (FADD) through the death domains (DD) of each protein. Subsequently, FADD
interacts with caspase-8 and/or -10 through the death effector domain (DED) of each
protein, resulting in the assembly of a death-inducing signaling complex (DISC). In the type
I pathway, extrinsic signals proteolytically activate caspase-8 and/or -10 followed by
stimulation of effector caspase-3 and -7, the key mediators of apoptosis, triggering
apoptosis. In the type II intrinsic pathway, however, apoptotic commitment requires an
amplification step involving the mitochondrial pathway triggered by caspase-8-dependent
cleavage of the Bid protein to its active form, t-Bid (Corallini et al., 2006; Hao et al., 2004;
Testa, 2010; Wang, 2008). Since TRAIL-R3, TRAIL-R4, and OPG lack intracellular functional
20 Advances in the Diagnosis of Coronary Atherosclerosis

portions, it is conceivable that they do not mediate apoptosis (Fig. 2.). However, even
binding of TRAIL to death receptors such as TRAIL-R1 or TRAIL-R2 can antagonize
apoptosis and induce cell proliferation under certain conditions in some cells. For example,
TRAIL-R1 and TRAIL-R2 recruit receptor-interacting protein (RIP) through their DD, which
can activate nuclear factor B (NFB). RIP-mediated NFB activation appears to induce cell
proliferation by TRAIL (Hao et al., 2004). TRAIL triggers recruitment of cellular FLICE-like
inhibitory protein (cFLIP) to DISC through their DED in some instances. Some cFLIPs
appear to prevent TRAIL-induced apoptosis (Hao et al., 2004; Wang, 2008). TRAIL
stimulation also recruits phosphoprotein enriched in diabetes (PED) to the DISC. This
process inhibits downstream caspase activation in some cases (Hao et al., 2004) (Fig. 2.).
Therefore, two contradictory steps should be considered when determining the biological
effects of TRAIL besides simple apoptotic signaling. First, TRAIL has death receptors
(TRAIL-R1 and TRAIL-R2) and antagonized decoy receptors (TRAIL-R3, TRAIL-R4, and
OPG) at the cell surface level. Second, these death receptors can counteract apoptotic
signaling at the intracellular level in certain contexts. Thus, TRAIL signaling is very
complicated.

Cysteine proteases

Transmembrane
Domain Receptor Binding Region

1 18 38 114 281

NH2 cleavage COOH

Cys 230

Soluble TRAIL

Zn

Formation of homotrimer
TRAIL consists of 281 amino acids with a transmembrane domain that separates the short intracellular
and extracellular domains. The extracellular domain of TRAIL is cleaved and released into the
bloodstream. TRAIL forms a homotrimer through interactions between zinc ions and each cysteine
residue.
Fig. 1. The Structure of TRAIL.
TRAIL has attracted clinical attention as a promising agent for the treatment of cancer since
it can induce apoptosis in various tumor cells without having any toxic effects on normal
cells (Finnberg and El-Deiry, 2008; Wang, 2008; Wu et al., 2004). Apart from high
expectations in the field of cancer, TRAIL also exhibits diverse biological effects on the
immune system, hematopoiesis, and metabolic disorders, including diabetes (Afford and
Multifunctional Role of TRAIL in Atherosclerosis and Cardiovascular Disease 21

Adams, 2005; Benito-Martin et al., 2009; Corallini et al., 2006; Testa, 2010; Vaccarezza et al.,
2007). Recently, the involvement of TRAIL in atherosclerosis and cardiovascular diseases
has been at the forefront of research efforts (Corallini et al., 2008; Kavurma and Bennett,
2008; Martin-Ventura et al., 2007). In this review, we focus on the role of TRAIL in the
cardiovascular system.

OPG

TRAIL-R1
TRAIL-R2 TRAIL-R3 TRAIL-R4
DD
DD
DD

FADD
DD

DD

RIP
cFLIP
DISC
PED
DED
DED

DED

DED

DED

Mitochondria
Type II
DED

DED

DED

Bid
Caspase-8, 10

Type I

Caspase-3, 7

apoptosis

Among the five known TRAIL receptors, TRAIL-R1 and –R2 can induce intracellular apoptotic signaling.
Upon binding of TRAIL to TRAIL-R1 or –R2, a death-inducing signaling complex (DISC) is formed
through the recruitment of Fas-associated death domain (FADD), and subsequently results in the
activation of caspase-8/10. In the type I intrinsic pathway, this activation is sufficient to conduct apoptosis
through downstream effector caspases. On the other hand, activated Bid by caspase-8 amplifies apoptotic
signaling through the mitochondria, which is a necessary step to induce effective downstream signaling in
the type II extrinsic pathway. In addition to this activation process, it appears that at least a few other
factors, such as receptor-interacting protein (RIP), cellular FLICE-like inhibitory protein (cFLIP), and
phosphoprotein enriched in diabetes (PED), are antagonistic to TRAIL-induced apoptosis.
Fig. 2. TRAIL receptors and downstream apoptotic signaling.

2. TRAIL and the cardiovascular system


TRAIL and its receptors are known to be expressed in the cardiovascular system. Therefore,
it is easily hypothesized that TRAIL systems may be involved in cardiovascular homeostasis
and disorders. To begin with, we will summarize several in vitro findings that demonstrate
22 Advances in the Diagnosis of Coronary Atherosclerosis

the direct effects of TRAIL on each cellular component in the arterial wall. We will then
discuss how TRAIL can act on the cardiovascular system based on evidence from animal
models. Subsequently, we try to make causal inferences relating to TRAIL’s role in
cardiovascular diseases from cross-sectional and longitudinal clinical studies in humans.
Finally, we report our recent clinical findings and debate whether TRAIL could be a
common biomarker that reflects early to advanced stage atherosclerotic changes.

2.1 Effects of TRAIL on cellular components in the arterial wall (in vitro findings)
Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are the principal cellular
components of the normal arterial wall. Immune-inflammatory infiltrates such as activated
T lymphocytes and macrophages play crucial roles in the onset and development of
atherosclerosis (Libby, 2002; Weyand et al., 2008). This process is triggered and exacerbated
by complicated interactions between modified lipoproteins and various cellular
components. Since TRAIL and its receptors are expressed in both physiological and
pathological arterial walls (Corallini et al., 2008; Kavurma and Bennett, 2008; Martin-
Ventura et al., 2007; Schoppet et al., 2004), it is hypothesized that TRAIL signaling could
regulate vascular wall homeostasis and morbid atherosclerosis. TRAIL is expressed in each
cellular component, both in resident cells (ECs, VSMCs) and in infiltrating cells (T
lymphocytes and macrophages) (Corallini et al., 2008; Kavurma and Bennett, 2008). To
determine the role of TRAIL in atherosclerosis, we must first determine the effects of TRAIL
on each cellular component based on results from in vitro studies.
ECs are located on the intima, which separates the blood stream from the vascular wall. In
addition, these cells have a critical function not only as a simple barrier but also as a
dynamic organ. Thus, ECs can regulate vascular tone, inflammation, thrombosis, and
vascular remodeling. Balanced homeostasis in ECs protects against atherosclerosis while
disruption of endothelial function results in the initiation and development of
atherosclerosis (Sandoo et al., 2010). Human ECs, both human umbilical vein ECs
(HUVECs) and human dermal microvascular ECs (HDMECs), express death receptors,
TRAIL-R1 and TRAIL-R2. As expected, TRAIL could induce apoptosis in ECs, although this
phenomenon was not observed in every cell (Li et al., 2003). This study also found that
apoptosis in ECs could be reduced by treatment with caspase inhibitors and by transfection
of dominant-negative FADD, suggesting the involvement of a FADD-caspase 8 pathway in
this apoptotic process (Li et al., 2003). Subsequently, Secchiero et al. showed that both
HUVECs and primary human aortic ECs express death TRAIL-R1, -R2 and decoy TRAIL-R3,
-R4. As predicted, the removal of serum and endothelial cell growth factor (tropic
withdrawal) from the cell culture system led to apoptosis. Surprisingly, TRAIL protected
HUVECs from tropic withdrawal-induced apoptosis. Activation of phosphatidylinositol 3-
kinase (PI3K)/Akt pathway by TRAIL appears to be involved in this anti-apoptotic action.
In addition to this unexpected anti-apoptotic property, TRAIL stimulated proliferation of
ECs in an ERK 1/2 dependent manner in this cell system (Secchiero et al., 2003). It has also
been reported that TRAIL alone can increase HUVEC cell proliferation (Alladina et al.,
2005). As reported above (Secchiero et al., 2003), inhibition of the PI3K/Akt pathway
sensitizes EC cells to TRAIL-induced apoptosis (Alladina et al., 2005). More detailed
investigations have found that inhibition of PI3K/Akt leads to activation of both extrinsic
and intrinsic apoptotic pathways and appears to sensitize HUVECs to TRAIL-induced
apoptosis by concurrent down-regulation of anti-apoptotic cFLIPs and Bcl-2 (Alladina et al.,
Multifunctional Role of TRAIL in Atherosclerosis and Cardiovascular Disease 23

2005). Taken together, these results suggest that the effects of TRAIL on ECs appear to
depend on cell culture conditions. Aside from cell apoptosis/proliferation, ECs have a
protective role against atherosclerosis by producing nitric oxide (NO), one of the most
important vasoactive factors. NO, a potent vasodilator, has antithrombotic and anti-
inflammatory activity (Sandoo et al., 2010). Interestingly, TRAIL stimulates NO production
through activation of endothelial NO synthase (eNOS) in HUVECs (Zauli et al., 2003). In
particular, eNOS trafficking appears to be involved in TRAIL-induced eNOS activation.
TRAIL can translocate eNOS from the cell membrane to the cytoplasm in HUVECs (Di
Pietro et al., 2006). However, microtubule disruption with nocodazole inhibits the eNOS
activity induced by TRAIL, suggesting that translocation of eNOS through cytoskeletal
reorganization may be necessary for TRAIL-induced NO production (Di Pietro et al., 2006).
NO synthesis with cytoskeletal alteration by TRAIL is possibly linked to the migration of
HUVECs stimulated by TRAIL treatment (Zauli et al., 2003).
In terms of atherosclerosis, attachment of blood leukocytes to ECs is recognized as the first
step in the initiation of this phenomenon (Libby, 2002). It is probable that TRAIL regulates
this process as well. An adhesion assay using co-cultured HUVECs and HL-60 leukocytes
showed that TRAIL increased the binding of leukocytes to ECs (Li et al., 2003). A later report
reproduced the pro-adhesive activity of TRAIL, although its effect was much less than that
of pro-inflammatory cytokines such as TNF and interleukin-1 (Secchiero et al., 2005).
When TRAIL and pro-inflammatory cytokines were evaluated at the same time, TRAIL pre-
treatment showed reverse inhibition of TNF and IL 1 induced HL-60 leukocyte
adhesion to HUVECs. This inhibitory effect of TRAIL on cell attachment was mediated by
down-regulation of the inflammatory chemokines CCL8 and CXCL10, which were
stimulated by TNF (Secchiero et al., 2005). In particular, TRAIL-R1 and TRAIL-R2
contributed to the TRAIL-induced down-regulation of chemokine release (Secchiero et al.,
2005). Again, TRAIL has multiple context-dependent effects. At any rate, TRAIL may play
an important role in EC function, both physiologically and pathologically.
Another physiological cellular component is the VSMCs, which are mainly localized in the
media and constitute the vascular structure. Once VSMCs are transformed, however, they
proliferate and migrate, contributing to the development of atherosclerosis (Ross, 1993). In
advanced atheromatous plaques, apoptosis of VSMCs in the fibrous cap may be involved in
plaque rupture. The former transformation exacerbates atherosclerosis (Libby, 2002),
whereas the latter leads to poorer outcome (Clarke et al., 2006). TRAIL participates in the
apoptosis and proliferation of VSMCs as well as ECs. For example, plaque-derived CD4 T
cells expressing TRAIL induce apoptosis of VSMCs, probably through the interaction of
TRAIL with TRAIL-R2 (Sato et al., 2006). On the other hand, TRAIL has been reported to act
on human and rat VSMCs as an anti-apoptotic factor through TRAIL-R1 and –R2 (Secchiero
et al., 2004). TRAIL promotes proliferation and migration of VSMCs through activation of
ERK (Secchiero et al., 2004). Furthermore, Kavurma et al. reported that TRAIL stimulated
proliferation of human VSMCs, although it induced apoptosis at high concentrations
(Kavurma et al., 2008). Interestingly, they showed the importance of insulin-like growth
factor-1 (IGF-1), one of the most potent growth factors, in TRAIL-induced proliferation of
VSMCs. Thus, TRAIL up-regulates expression of the IGF-1 receptor in an NFB-dependent
manner (Kavurma et al., 2008). It is thus too soon to decide whether TRAIL causes apoptosis
or proliferation in VSMCs, even in vitro.
In addition to the seemingly contradictory findings in ECs and VSMCs, a few reports have
suggested that TRAIL has a pro-apoptotic effect on inflammatory lymphocytes (Janssen et
24 Advances in the Diagnosis of Coronary Atherosclerosis

al., 2005) and macrophages (Kaplan et al., 2000). In either case, TRAIL appears to have
multiple functions in both physiological and pathological cells under various conditions. As
described previously, the existence of five different types of TRAIL receptors and the cross-
talk among multiple post-receptor signaling pathways may explain these diverse effects. On
the basis of in vitro findings, it is very difficult to speculate the impact of TRAIL on
atherosclerotic lesions in vivo, although the in vitro findings clearly indicate the involvement
of the TRAIL/TRAIL receptor system in atherogenesis. In the next section, we try
summarizing and discussing whether TRAIL protects against or exacerbates atherosclerosis
in ways aside from its function as a mere pro-apoptotic factor.

2.2 Role of TRAIL in atherosclerosis and vascular injury (in vivo findings)
With regard to the role of TRAIL in the vascular wall, Secchiero et al. first demonstrated
compelling in vivo findings using diabetic apolipoprotein E (apoE)-null mice that mimic the
atherosclerotic lesions observed in humans (Secchiero et al., 2006). That is, intraperitoneal
administration of recombinant human TRAIL into these mice resulted in a transient high
concentration of TRAIL and subsequent protection against the development of
atherosclerosis (Secchiero et al., 2006). Secchiero et al. carefully investigated the effects of
TRAIL on in vivo atherosclerotic lesions using an adeno-associated virus containing TRAIL
to ensure a low but sustained expression of TRAIL, similar to physiological conditions.
TRAIL again attenuated the development of atherosclerotic plaques, even under these
conditions (Secchiero et al., 2006). One of the mechanisms responsible for this effect may be
selective apoptosis of infiltrating macrophages in plaque lesions. At the same time,
increased VSMC levels were also observed in the fibrous caps of the atherosclerotic lesions.
This increase may contribute to stabilization of the atherosclerotic plaques (Secchiero et al.,
2006). Therefore, TRAIL appears to act as a protective factor against atherosclerosis in vivo.
Subsequently, Kavurma’s group showed the direct effects of TRAIL on VSMCs in vivo. They
have already reported that TRAIL stimulates VSMC proliferation in vitro (Kavurma et al.,
2008). To prove this using an in vivo model, they used a cuff-induced vascular injury method
in TRAIL-null mice. In wild-type mice, this procedure can induce VSMC proliferation and
intimal thickening in response to vascular injury. However, TRAIL-null mice were protected
from neointimal formation and displayed reduced VSMC proliferation, suggesting a
significant role for TRAIL in VSMC proliferation in vivo (Chan et al., 2010). Although this
report showed the direct effects of TRAIL on VSMCs, the lack of atherogenic factors
involved in the formation of vascular lesions should be studied further. Very recently, it has
been reported that TRAIL attenuates the development of atherosclerosis using TRAIL
(TRAIL −/−)/apoE (apoE −/−) double-knockout mice (Watt et al., 2011). TRAIL −/− apoE
−/− mice had significantly larger atheromatous lesions compared with apoE −/− control
mice at 8 weeks. The larger lesions in TRAIL −/− apoE −/− mice appeared to be due to an
increase in the number of lesional VSMCs, suggesting the anti-atherogenic action of TRAIL.
Intriguingly, the difference in atheromatous lesion size among these mice became smaller at
12 weeks. In contrast to Secchiero’s findings, the lack of TRAIL had no effect on the
macrophage content in the atheromatous lesions. There are still unsolved and controversial
issues with regard to the precise mechanisms by which TRAIL acts on the arterial wall.
However, TRAIL at least appears to protect against atherogenic lesions as a whole in vivo.

2.3 TRAIL and cardiovascular disease and prognosis (clinical findings)


Clinical studies of the relationship between TRAIL and cardiovascular diseases are also of
interest. Recently, a significant relationship has been reported by a number of studies. The
Multifunctional Role of TRAIL in Atherosclerosis and Cardiovascular Disease 25

first study demonstrated that serum TRAIL levels were significantly lower in patients with
acute coronary syndrome (ACS) compared with those with stable angina and normal
coronary arteries (Michowitz et al., 2005). Subsequently, Schoppet et al. found a tendency
for serum TRAIL levels to be lower in patients with coronary artery disease (CAD)
compared with subjects without CAD, although this difference was not significant. To
further investigate this relationship, we examined serum TRAIL levels in 285 subjects who
underwent coronary angiography for suspected CAD. Interestingly, we found that serum
TRAIL levels were inversely associated with the severity of CAD (Mori et al., 2010). In
particular, TRAIL levels in patients with severe three-vessel disease (VD) were significantly
lower than in those without CAD (Mori et al., 2010). Moreover, TRAIL was an independent
and negative contributor for the presence of CAD (Mori et al., 2010). Taken together, these
results suggest that lower TRAIL levels may reflect the advancement of CAD.
In addition to cross-sectional findings, recent studies have also suggested that TRAIL can be
a protective predictor against cardiovascular prognosis. As previously reported (Michowitz
et al., 2005), Secchiero et al. showed that serum TRAIL levels were significantly lower in
patients with acute myocardial infarction (AMI) at baseline (within 24 hours from
admission) compared with healthy subjects (Secchiero et al., 2009). Interestingly, serum
TRAIL levels at baseline were significantly lower in patients with in-hospital adverse events
compared with those who did not experience these events (Secchiero et al., 2009).
Subsequently, they observed that serum TRAIL levels in AMI patients gradually recovered
at discharge. Furthermore, low TRAIL levels at discharge were associated with an increased
incidence of cardiac death and heart failure in the 12-month follow-up (Secchiero et al.,
2009). The prognostic value of TRAIL was also examined in 351 patients with advanced
heart failure (HF). Again, low serum TRAIL levels were related to a worse prognosis. The
risk of mortality dropped by 70% in the highest quartile of TRAIL levels, suggesting that
TRAIL is a strong inverse predictor of mortality in patients with advanced HF (Niessner et
al., 2009). In addition, the role of TRAIL as a more general predictor of mortality and not
limited to patients with AMI or advanced HF was investigated by a large prospective
population-based study of older people (Volpato et al., 2011). Baseline TRAIL levels were
inversely related to all-cause mortality over a period of six years (Volpato et al., 2011). As
expected, more detailed analyses revealed that the prognostic effect of TRAIL levels was
strong and highly significant in subjects with prevalent cardiovascular diseases (Volpato et
al., 2011). These findings expand on the predictive ability of TRAIL at a population level
with a longer follow-up period. Moreover, Secchiero et al. focused not only on TRAIL but
also on OPG. As was described earlier, OPG is a soluble neutralizing receptor for TRAIL. In
contrast to TRAIL, it is known that serum OPG levels are positively associated with the
presence and severity of CAD and are inversely correlated with prognosis (Abedin et al.,
2007; Jono et al., 2002; Kiechl et al., 2004; Omland et al., 2008; Rhee et al., 2005; Schoppet et
al., 2003). In this context, both TRAIL and OPG were evaluated at the same time. The
researchers found that the OPG/TRAIL ratio was significantly higher in patients with acute
AMI who developed HF during the follow-up period (Secchiero et al., 2010). The increase in
OPG, which is a decoy TRAIL receptor, may act against TRAIL and thus negate its
protective effects on the cardiovascular system. Thus, an unbalanced OPG/TRAIL ratio may
be a more accurate predictor of prognosis after AMI.

2.4 Modulating factors in TRAIL-associated biological and clinical effects


Several reports suggest that expression and/or serum levels of TRAIL are modulated by
certain factors. As described above, serum TRAIL levels drop after AMI. Since proteolytic
26 Advances in the Diagnosis of Coronary Atherosclerosis

enzymes including matrix metalloproteinases (MMPs) are released following AMI,


researchers have investigated whether MMPs could cleave TRAIL and thus decrease TRAIL
levels, inducing AMI. In addition to MMPs, Secchiero et al. have simultaneously measured
tissue inhibitors of MMPs (TIMPs) that antagonize MMPs (Secchiero et al., 2010). Among the
examined MMPs and TIMPs, the circulating MMP2/TIMP2 ratio showed a significant
inverse correlation with serum TRAIL levels in AMI patients (Secchiero et al., 2010). Indeed,
MMP2 cleaved TRAIL and abrogated its biological activity in vitro (Secchiero et al., 2010).
Therefore, an elevated MMP2/TIMP2 ratio following AMI may cause degradation of
TRAIL, resulting in a poorer outcome. With regards to TRAIL expression, insulin has been
reported to down-regulate TRAIL expression in VSMCs both in vitro and in vivo (Corallini et
al., 2007). Since VSMCs are also known to release bioactive NO in response to TRAIL, it is
hypothesized that chronic insulin exposure in VSMCs may induce vascular dysfunction
through TRAIL suppression in diabetic patients with hyperinsulinemia or those undergoing
insulin treatment. Moreover, it has been reported that activated protein C (APC), an
antithrombotic and anti-inflammatory serine protease, inhibits TRAIL expression in
HUVECs (O’Brien et al., 2007). In that study, intracellular signaling was thoroughly
examined (O’Brien et al., 2007). However, there has been some criticism of APC-mediated
decrease of TRAIL levels. It is currently unknown whether this reflects a decrease in cell-
surface TRAIL expression or of TRAIL release from its intracellular pool (Secchiero and
Zauli, 2008).
Statins also appear to down-regulate TRAIL expression in cytotoxic CD4 T cells in patients
with ACS, resulting in protection against destabilization of plaques (Sato et al., 2010). That
is, CD4 T cells are enriched in the blood of ACS patients and induce strong apoptosis of ECs,
probably through TRAIL-R2 (DR5) (Sato et al., 2010). This EC apoptosis may be involved in
the erosive progression of vulnerable plaques. Interestingly, statins directly block CD4 T
cell-mediated EC apoptosis (Sato et al., 2010). Therefore, this protection against endothelial
injury shown by statins may explain one of the pleiotropic effects of statins in cardiovascular
events. On the other hand, it has been reported that statins augment TRAIL-induced
apoptosis in tumor cells but not in normal cells (Jin et al., 2002), although this is not the case
in vascular cells. In this regard, pioglitazone, an anti-diabetic agent, also enhances TRAIL-
induced apoptosis in tumor cells (Goke et al., 2000). In clinical studies, use of various
medications against diabetes, hypertension, dyslipidemia, and so on is often unavoidable.
To interpret the association between TRAIL and cardiovascular lesions, it is important to
pay attention to the types of interventions used.

2.5 TRAIL as a possible biomarker in early atherosclerotic lesions


As noted above, lower TRAIL levels appear to be correlated with more severe
cardiovascular lesions and poorer prognosis. Thus, the next question is whether TRAIL
levels also inversely reflect early atherosclerotic lesions. We therefore examined the
association between TRAIL and intima-media thickness (IMT), which is a surrogate marker
for atherosclerotic changes, using ultrasonography in 416 diabetic patients. We found no
significant association between serum TRAIL levels and IMT (Kawano et al., 2011).
Interestingly, when we focused on subjects with macrovascular diseases such as CAD,
cerebrovascular diseases, and arteriosclerosis obliterans, there was a significant and
reproducible association between TRAIL levels and carotid IMT. These findings suggest that
TRAIL may not be a good candidate biomarker for early atherosclerotic lesions. However,
TRAIL still appears to be a good biomarker of advanced atheroscletoic lesions.
Multifunctional Role of TRAIL in Atherosclerosis and Cardiovascular Disease 27

In contrast to IMT, which indicates morphological wall thickening, endothelial dysfunction


is recognized as reflecting earlier functional vascular damage. Flow-mediated dilatation
(FMD) is a representative method of non-invasive evaluation of endothelial function. This
ultrasound-based method quantifies vasodilatation in response to NO production from
endothelial cells induced by shear stress (Corretti et al., 2002; Patel and Celermajer, 2006; Ter
Avest et al., 2007). Since in vitro findings have suggested various biological effects of TRAIL
on endothelial cells (as described previously), investigation of the association between
TRAIL and endothelial function is of interest. In this context, we tried measuring FMD and
TRAIL levels in 109 subjects (57 men and 52 women, aged 48.4 ± 16.6 years). In the trial, we
focused on the following two points: First, we targeted healthy subjects to avoid various
confounding biases such as metabolic disorders with concomitant drug intervention.
Second, we made sure of the accuracy of FMD. It is often noted that the value of FMD is
highly dependent on the technique used by various institutions (Ter Avest et al., 2007). Very
recently, semi-automated equipment (Unex Co. Ltd., Nagoya, Japan) has become available
for evaluating FMD that gives good reproducibility (Tomiyama et al., 2008; Tomiyama and
Yamashina, 2010). We used this new equipment in this study. The serum TRAIL level was
measured by an enzyme-linked immunosorbent assay kit (R&D systems, Minneapolis,
USA). The mean serum TRAIL level was 75.2 ± 20.7 pg/ml, with a range of 32.4–147.4
pg/ml. The TRAIL level was not significantly correlated with FMD (ρ = −0.128, p = 0.184)
(Fig. 3) (unpublished data). Again, these findings suggest that TRAIL may not be a good
candidate as a biomarker of early atherosclerotic lesions.

Fig. 3. The association of TRAIL with endothelial function in 109 healthy subjects.
Flow-mediated dilatation (FMD) was used to evaluate endothelial function in 109 healthy
subjects. There was no correlation between serum TRAIL levels and FMD (ρ = −0.128, p = 0.148).
28 Advances in the Diagnosis of Coronary Atherosclerosis

3. Conclusions
Recent emerging evidence has suggested the definite involvement of TRAIL in
cardiovascular diseases. Taken together, these results have shown that lower serum TRAIL
levels appear to be associated with worse prognosis in patients with CAD and HF. One of
most important question is why TRAIL levels are lower in such conditions. It is necessary to
investigate how TRAIL is produced and cleared in humans in future studies. In addition, to
confirm the direct effects of TRAIL on cardiovascular diseases, administration of
recombinant TRAIL may be a powerful approach. In fact, recombinant TRAIL induces
apoptosis in a wide variety of tumor cells and prevents tumor progression and metastasis in
the field of cancer therapy (Johnstone et al., 2008; Wang, 2008; Wu et al., 2004). Further
studies are needed to address these problems.

4. Acknowledgment
This review was supported in part by a Grant-in-Aid for scientific research (No. 20591068)
from the Japan Society for the Promotion of Science (to ME and KM). The authors thank Dr.
Naoya Kawano, Dr. Hidenori Koyama, Ms. Yuko Kikukawa, Ms. Hisako Fujii, Dr. Sanae
Fukuda, Mr. Hidekichi Tokai, Dr. Yoshinobu Hirayama, and Dr. Yasuyoshi Watanabe for
their help with the study to investigate the association between TRAIL levels and FMD in
healthy subjects (Fig. 3).

5. References
Abedin, M, et al. (2007). Relation of osteoprotegerin to coronary calcium and aortic plaque
(from the Dallas Heart Study). Am J Cardiol, Vol. 99, No. 4, (February 2007) pp. 513-
518, ISSN 0002-9149
Afford, SC, Adams, DH. (2005). Following the TRAIL from hepatitis C virus and alcohol to
fatty liver. Gut, Vol. 54, No. 11, (October 2005) pp. 1518-1520, ISSN 0017-5749
Alladina, SJ, et al. (2005). TRAIL-induced apoptosis in human vascular endothelium is
regulated by phosphatidylinositol 3-kinase/Akt through the short form of cellular
FLIP and Bcl-2. J Vasc Res, Vol. 42, No. 4, (June 2005) pp. 337-47, ISSN 1018-1172
Benito-Martin, A, et al. (2009). Transcriptomics illustrate a deadly TRAIL to diabetic
nephropathy. Nefrologia, Vol. 29, No. 1, (February 2009) pp. 13-19, ISSN 0211-6995
Chan, J, et al. (2010). TRAIL promotes VSMC proliferation and neointima formation in a
FGF-2-, Sp1 phosphorylation-, and NFkappaB-dependent manner. Circ Res, Vol.
106, No. 6, (February 2010) pp. 1061-71, ISSN 1524-4571
Clarke, MC, et al. (2006). Apoptosis of vascular smooth muscle cells induces features of
plaque vulnerability in atherosclerosis. Nat Med, Vol. 12, No. 9, (August 2006) pp.
1075-1080, ISSN 1078-8956
Corallini, F, et al. (2006). TRAIL, caspases and maturation of normal and leukemic myeloid
precursors. Leuk Lymphoma, Vol. 47, No. 8, (September 2006) pp. 1459-1468, ISSN
1042-8194
Corallini, F, et al. (2007). Insulin down-regulates TRAIL expression in vascular smooth
muscle cells both in vivo and in vitro. J Cell Physiol, Vol. 212, No. 1, (March 2007)
pp. 89-95, ISSN 0021-9541
Multifunctional Role of TRAIL in Atherosclerosis and Cardiovascular Disease 29

Corallini, F, et al. (2008). TRAIL and osteoprotegerin: a role in endothelial physiopathology?


Front Biosci, Vol. 13, No. (November 2007) pp. 135-147, ISSN 1093-4715
Corretti, MC, et al. (2002). Guidelines for the ultrasound assessment of endothelial-
dependent flow-mediated vasodilation of the brachial artery: a report of the
International Brachial Artery Reactivity Task Force. J Am Coll Cardiol, Vol. 39, No. 2,
(January 2002) pp. 257-265, ISSN 0735-1097
Di Pietro, R, et al. (2006). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)
regulates endothelial nitric oxide synthase (eNOS) activity and its localization
within the human vein endothelial cells (HUVEC) in culture. J Cell Biochem, Vol. 97,
No. 4, (October 2005) pp. 782-794, ISSN 0730-2312
Finnberg, N, El-Deiry, WS. (2008). TRAIL death receptors as tumor suppressors and drug
targets. Cell Cycle, Vol. 7, No. 11, (May 2008) pp. 1525-1528, ISSN 1551-4005
Goke, R, et al. (2000). Regulation of TRAIL-induced apoptosis by transcription factors. Cell
Immunol, Vol. 201, No. 2, (June 2000) pp. 77-82, ISSN 0008-8749
Hao, C, et al. (2004). Modulation of TRAIL signaling complex. Vitam Horm, Vol. 67, No.
(April 2004) pp. 81-99, ISSN 0083-6729
Hymowitz, SG, et al. (1999). Triggering cell death: the crystal structure of Apo2L/TRAIL in
a complex with death receptor 5. Mol Cell, Vol. 4, No. 4, (November 1999) pp. 563-
571, ISSN 1097-2765
Janssen, EM, et al. (2005). CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-
mediated activation-induced cell death. Nature, Vol. 434, No. 7029, (March 2005)
pp. 88-93, ISSN 1476-4687
Jin, Z, et al. (2002). Enhanced sensitivity of G1 arrested human cancer cells suggests a novel
therapeutic strategy using a combination of simvastatin and TRAIL. Cell Cycle, Vol.
1, No. 1, (November 2002) pp. 82-89, ISSN 1538-4101
Johnstone, RW, et al. (2008). The TRAIL apoptotic pathway in cancer onset, progression and
therapy. Nat Rev Cancer, Vol. 8, No. 10, (September 2008) pp. 782-798, ISSN 1474-
1768
Jono, S, et al. (2002). Serum osteoprotegerin levels are associated with the presence and
severity of coronary artery disease. Circulation, Vol. 106, No. 10, (September 2002)
pp. 1192-1194, ISSN 1524-4539
Kaplan, MJ, et al. (2000). TRAIL (Apo2 ligand) and TWEAK (Apo3 ligand) mediate CD4+ T
cell killing of antigen-presenting macrophages. J Immunol, Vol. 164, No. 6, (March
2000) pp. 2897-2904, ISSN 0022-1767
Kavurma, MM, Bennett, MR. (2008). Expression, regulation and function of trail in
atherosclerosis. Biochem Pharmacol, Vol. 75, No. 7, (December 2007) pp. 1441-1450,
ISSN 1873-2968
Kavurma, MM, et al. (2008). TRAIL stimulates proliferation of vascular smooth muscle cells
via activation of NF-kappaB and induction of insulin-like growth factor-1 receptor.
J Biol Chem, Vol. 283, No. 12, (January 2008) pp. 7754-7762, ISSN 0021-9258
Kawano, N, et al. (2011). Association of serum TRAIL levels with atherosclerosis in patients
with type 2 diabetes mellitus. Diabetes Res Clin Pract, Vol. 91, No. 3, (January 2011)
pp. 316-320, ISSN 1872-8227
Kiechl, S, et al. (2004). Osteoprotegerin is a risk factor for progressive atherosclerosis and
cardiovascular disease. Circulation, Vol. 109, No. 18, (May 2004) pp. 2175-2180,
ISSN 1524-4539
30 Advances in the Diagnosis of Coronary Atherosclerosis

Li, JH, et al. (2003). TRAIL induces apoptosis and inflammatory gene expression in human
endothelial cells. J Immunol, Vol. 171, No. 3, (July 2003) pp. 1526-1533, ISSN 0022-
1767
Libby, P. (2002). Inflammation in atherosclerosis. Nature, Vol. 420, No. 6917, (December
2002) pp. 868-874, ISSN 0028-0836
Martin-Ventura, JL, et al. (2007). Trail and vascular injury. Front Biosci, Vol. 12, No. (May
2007) pp. 3656-3667, ISSN 1093-4715
Michowitz, Y, et al. (2005). The involvement of tumor necrosis factor-related apoptosis-
inducing ligand (TRAIL) in atherosclerosis. J Am Coll Cardiol, Vol. 45, No. 7, (April
2005) pp. 1018-1024, ISSN 0735-1097
Mori, K, et al. (2010). Association of serum TRAIL level with coronary artery disease. Thromb
Res, Vol. 125, No. 4, (December 2009) pp. 322-325, ISSN 1879-2472
Niessner, A, et al. (2009). Prognostic value of apoptosis markers in advanced heart failure
patients. Eur Heart J, Vol. 30, No. 7, (February 2009) pp. 789-796, ISSN 1522-9645
O'Brien, LA, et al. (2007). Activated protein C decreases tumor necrosis factor related
apoptosis-inducing ligand by an EPCR- independent mechanism involving Egr-
1/Erk-1/2 activation. Arterioscler Thromb Vasc Biol, Vol. 27, No. 12, (October 2007)
pp. 2634-2641, ISSN 1524-4636
Omland, T, et al. (2008). Circulating osteoprotegerin levels and long-term prognosis in
patients with acute coronary syndromes. J Am Coll Cardiol, Vol. 51, No. 6, (February
2008) pp. 627-633, ISSN 1558-3597
Patel, S, Celermajer, DS. (2006). Assessment of vascular disease using arterial flow mediated
dilatation. Pharmacol Rep, Vol. 58 Suppl, No. (March 2007) pp. 3-7, ISSN 1734-1140
Pitti, RM, et al. (1996). Induction of apoptosis by Apo-2 ligand, a new member of the tumor
necrosis factor cytokine family. J Biol Chem, Vol. 271, No. 22, (May 1996) pp. 12687-
12690, ISSN 0021-9258
Rhee, EJ, et al. (2005). Relationship of serum osteoprotegerin levels with coronary artery
disease severity, left ventricular hypertrophy and C-reactive protein. Clin Sci
(Lond), Vol. 108, No. 3, (December 2004) pp. 237-243, ISSN 0143-5221
Ross, R. (1993). The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature, Vol.
362, No. 6423, (April 1993) pp. 801-809, ISSN 0028-0836
Sandoo, A, et al. (2010). The endothelium and its role in regulating vascular tone. Open
Cardiovasc Med J, Vol. 4, No. (February 2011) pp. 302-12, ISSN 1874-1924 (Electronic)
Sato, K, et al. (2006). TRAIL-expressing T cells induce apoptosis of vascular smooth muscle
cells in the atherosclerotic plaque. J Exp Med, Vol. 203, No. 1, (January 2006) pp.
239-250, ISSN 0022-1007
Sato, K, et al. (2010). Statins reduce endothelial cell apoptosis via inhibition of TRAIL
expression on activated CD4 T cells in acute coronary syndrome. Atherosclerosis,
Vol. 213, No. 1, (May 2010) pp. 33-39, ISSN 1879-1484
Schoppet, M, et al. (2003). Increased osteoprotegerin serum levels in men with coronary
artery disease. J Clin Endocrinol Metab, Vol. 88, No. 3, (March 2003) pp. 1024-1028,
ISSN 0021-972X
Schoppet, M, et al. (2004). Localization of osteoprotegerin, tumor necrosis factor-related
apoptosis-inducing ligand, and receptor activator of nuclear factor-kappaB ligand
in Monckeberg's sclerosis and atherosclerosis. J Clin Endocrinol Metab, Vol. 89, No.
8, (August 2004) pp. 4104-4112, ISSN 0021-972X
Multifunctional Role of TRAIL in Atherosclerosis and Cardiovascular Disease 31

Secchiero, P, et al. (2003). TRAIL promotes the survival and proliferation of primary human
vascular endothelial cells by activating the Akt and ERK pathways. Circulation, Vol.
107, No. 17, (April 2003) pp. 2250-2256, ISSN 1524-4539
Secchiero, P, et al. (2004). TRAIL promotes the survival, migration and proliferation of
vascular smooth muscle cells. Cell Mol Life Sci, Vol. 61, No. 15, (August 2004) pp.
1965-1974, ISSN 1420-682X
Secchiero, P, et al. (2005). TRAIL counteracts the proadhesive activity of inflammatory
cytokines in endothelial cells by down-modulating CCL8 and CXCL10 chemokine
expression and release. Blood, Vol. 105, No. 9, (January 2005) pp. 3413-3419, ISSN
0006-4971
Secchiero, P, et al. (2006). Systemic tumor necrosis factor-related apoptosis-inducing ligand
delivery shows antiatherosclerotic activity in apolipoprotein E-null diabetic mice.
Circulation, Vol. 114, No. 14, (September 2006) pp. 1522-1530, ISSN 1524-4539
Secchiero, P, Zauli, G. (2008). The puzzling role of TRAIL in endothelial cell biology.
Arterioscler Thromb Vasc Biol, Vol. 28, No. 2, (January 2008) pp. e4; author reply e5-6,
ISSN 1524-4636
Secchiero, P, et al. (2009). Potential prognostic significance of decreased serum levels of
TRAIL after acute myocardial infarction. PLoS One, Vol. 4, No. 2, (February 2009)
pp. e4442, ISSN 1932-6203
Secchiero, P, et al. (2010). An imbalanced OPG/TRAIL ratio is associated to severe acute
myocardial infarction. Atherosclerosis, Vol. 210, No. 1, (December 2009) pp. 274-277,
ISSN 1879-1484
Secchiero, P, et al. (2010). Metalloproteinase 2 cleaves in vitro recombinant TRAIL: potential
implications for the decreased serum levels of TRAIL after acute myocardial
infarction. Atherosclerosis, Vol. 211, No. 1, (March 2010) pp. 333-336, ISSN 1879-1484
Ter Avest, E, et al. (2007). What is the role of non-invasive measurements of atherosclerosis
in individual cardiovascular risk prediction? Clin Sci (Lond), Vol. 112, No. 10, (April
2007) pp. 507-516, ISSN 1470-8736
Testa, U. (2010). TRAIL/TRAIL-R in hematologic malignancies. J Cell Biochem, Vol. 110, No.
1, (March 2010) pp. 21-34, ISSN 1097-4644
Tomiyama, H, et al. (2008). The relationships of cardiovascular disease risk factors to flow-
mediated dilatation in Japanese subjects free of cardiovascular disease. Hypertens
Res, Vol. 31, No. 11, (December 2008) pp. 2019-2025, ISSN 0916-9636
Tomiyama, H, Yamashina, A. (2010). Non-invasive vascular function tests: their
pathophysiological background and clinical application. Circ J, Vol. 74, No. 1,
(November 2009) pp. 24-33, ISSN 1347-4820
Vaccarezza, M, et al. (2007). A role of the TRAIL-TRAIL receptor system in the pathogenesis
of diabetes. Acta Biomed, Vol. 78 Suppl 1, No. (May 2007) pp. 262-267, ISSN 0392-
4203
Venuraju, SM, et al. (2010). Osteoprotegerin as a predictor of coronary artery disease and
cardiovascular mortality and morbidity. J Am Coll Cardiol, Vol. 55, No. 19, (May
2010) pp. 2049-2061, ISSN 1558-3597
Volpato, S, et al. (2011). Association of tumor necrosis factor-related apoptosis-inducing
ligand with total and cardiovascular mortality in older adults. Atherosclerosis, Vol.
215, No. 2. (December 2010) pp. 452-458, ISSN 1879-1484
32 Advances in the Diagnosis of Coronary Atherosclerosis

Wang, S. (2008). The promise of cancer therapeutics targeting the TNF-related apoptosis-
inducing ligand and TRAIL receptor pathway. Oncogene, Vol. 27, No. 48, (October
2008) pp. 6207-6215, ISSN 1476-5594
Watt, V, et al. (2011). TRAIL attenuates the development of atherosclerosis in apolipoprotein
E deficient mice. Atherosclerosis, Vol. No. (February 2011) pp. ISSN 1879-1484
Weyand, CM, et al. (2008). T cells in arteritis and atherosclerosis. Curr Opin Lipidol, Vol. 19,
No. 5, (October 2008) pp. 469-477, ISSN 0957-9672
Wiley, SR, et al. (1995). Identification and characterization of a new member of the TNF
family that induces apoptosis. Immunity, Vol. 3, No. 6, (December 1995) pp. 673-
682, ISSN 1074-7613
Wu, XX, et al. (2004). TRAIL and chemotherapeutic drugs in cancer therapy. Vitam Horm,
Vol. 67, No. (April 2004) pp. 365-383, ISSN 0083-6729
Zauli, G, et al. (2003). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)
sequentially upregulates nitric oxide and prostanoid production in primary human
endothelial cells. Circ Res, Vol. 92, No. 7, (March 2003) pp. 732-740, ISSN 1524-4571
3

Indications for Coronary Angiography


Karl Poon and Darren Walters
The Prince Charles Hospital
Australia

1. Introduction
Since the first serendipitous selective coronary angiography was performed by pediatric
cardiologist Dr. Mason Sones in 1958 during ventriculography and aortography (Sones et
al., 1959), invasive coronary angiography has become well established as the diagnostic gold
standard for coronary artery disease.
Coronary angiography provides definitive information of “luminology” (radiographic
outline using a contrast agent) of the coronary arteries. It is the standard with which other
diagnostic modalities are compared. Millions are performed annually worldwide. Whilst the
risk of coronary angiography of complications is very low in experienced hands the
procedure is invasive and, requires dedicated infrastructure and expense. There are certain
specific indications for coronary angiography. The general indication for a coronary
angiography is to define the coronary artery anatomy. More detailed guidelines for
coronary angiography date back to 1999 from the American College of Cardiology (Scanlon
et al., 1999). This of course predated the enormous technological advancement over the past
decade, the development in computed tomography coronary angiography, the widespread
adoption of coronary stenting, the improvement afforded by drug eluting stents, and the
availability of new antiplatelet therapy that enhanced the safety of all coronary
interventions.
This chapter examines the appropriate indications for invasive coronary angiography in the
current era, assesses the role of non invasive CT coronary angiography, and incorporates the
emerging adjunctive role in the management of patients undergoing percutaneous cardiac
structural interventions. Case vignettes are included in this chapter to illustrate the
indications and use of coronary angiography in the current era.
It must be stressed whilst guidelines do provide recommendations regarding the indications
for invasive coronary angiography, many more complex clinical and non clinical factors
exist for each individual patient and the clinician must exercise clinical acumen to proceed
to perform an invasive diagnostic and/or therapeutic procedure.

2. Indications for coronary angiography


Coronary angiography involves the opacification of the lumen in coronary arteries and
acquisition of this luminogram under fluoroscopy. Consequently the indication for coronary
angiography is predominantly focused on the diagnosis of any conditions that can lead to
luminal compromise. This anatomical study defines the origin, course and pattern of
34 Advances in the Diagnosis of Coronary Atherosclerosis

epicardial arteries, with a spatial resolution that is still superior to other non-invasive
imaging modalities. There are however limitations to coronary angiography. Aside from the
obvious invasive nature and attendant risks, it provides only a projection of the lumen
outlined by contrast and hence no information on the extent of atheroma contained within
the vessel wall or the functional significance of the stenosis which can only be inferred. Both
of these limitations can be overcome by the use of intravascular modalities such as
intravascular ultrasound, optical coherence tomography and fractional flow reserve
assessment. When combined with these adjuncts, coronary angiography serves as a
formidable tool in guiding revascularization strategies, with a body of scientific data that
has been well validated to provide long term prognostic benefit.
The guiding principle in determining the indication behind the decision to undertake
coronary angiography in a given patients is similar to that underpinning the decision to
undertake any invasive investigations in medicine. The decision to proceed is based on that
the knowledge gained from the investigation outweighs the perceived risk, that the result
will benefit the patient, in that it will likely change management and the available treatment
option will improve symptoms and/or prognosis. The risks and complications of coronary
angiography are covered elsewhere in this book and this chapter will focus on the
indications of coronary angiography. Relative and absolute contraindications are also
covered briefly.
The indications for coronary angiography have been divided into the follow clinical
categories:
1. Coronary artery disease
1. Stable pattern
i. suspected coronary artery disease
ii. known coronary artery disease
2. Unstable pattern
i. Acute coronary syndrome with unstable haemodynamics or rhythm
ii. Unstable angina and Non-ST elevation MI
iii. ST elevation MI
iv. Out of hospital cardiac arrest
3. Special considerations
i. Congestive cardiac failure
ii. Preoperative assessment for non cardiac surgery
iii. The value of non invasive computed tomography coronary angiography
4. Relative and absolute contraindications
2. Prelude to structural cardiac disease/valvular heart disease intervention
3. Case vignettes of the use of coronary angiography in the contemporary era

3. Coronary artery disease


Coronary angiography is pivotal in the diagnostic algorithm as the gold standard for the
confirmation of coronary artery stenosis. A number of well defined clinical scenarios
often lead to the suspicion of coronary artery disease. This section is separated into
patients who present in stable pattern, symptomatic or asymptomatic; and patients who
present in an acute manner highly suggestive of coronary artery disease, i.e. acute
coronary syndromes. Specifics scenarios, such as the role of coronary angiography in the
Indications for Coronary Angiography 35

investigation of congestive cardiac failure, pre-valvular surgery and the emerging role of
computed tomography coronary angiography will be covered too.

3.1.1 Coronary artery disease – stable pattern – suspected coronary artery disease
The assessment of patients with symptoms suggestive of coronary artery disease (CAD)
depends on three key factors: the clinical assessment of the quality of symptoms, the clinical
risk profile, and the presence of existing abnormalities on surface electrocardiogram
suggestive of CAD.
The ACC/AHA guidelines on exercise testing (Gibbons, 2002) provide classification of
symptoms as typical, atypical angina and non-anginal chest pain (table 1). It is well known
symptoms alone have a poor predictive value and differ between males and females
(Hemingway et al, 2006, 2008). Combined with the underlying clinical risk profile however,
a more reliable pre-test probability on CAD can be generated (table 2).

Chest pain Definition


Typical/definite angina Substernal chest pain or equivalent
 Provoked by exertion or emotional stress
 Relieved by rest/nitroglycerin
Atypical/probable Chest pain or discomfort with two characteristics of definite or
angina typical angina
Non-anginal chest pain Chest pain or discomfort that meets one or none of the typical
angina characteristics

Table 1. Classification of chest pain

Age Sex Typical/definite Atypical/probable Nonanginal Asymptomatic


angina angina chest pain
<39 Male Intermediate Intermediate Low Very low
Female Intermediate Very low Very low Very low
40- Male High Intermediate Intermediate Low
49 Female Intermediate Low Very low Very low
50- Male High Intermediate Intermediate Low
59 Female Intermediate Intermediate Low Very low
>60 Male High Intermediate Intermediate Low
Female High Intermediate Intermediate Low

Table 2. Pretest probability of coronary artery disease based on age, sex and symptoms
(adapted from Gibbons et al, 2002)
The resting surface electrocardiogram (ECG) is important in the initial evaluation of the
patient for two reasons. First, it provides a screening tool to assess for remote infarctions
(Ammar et al 2004; Michael et al, 2007), with changes such as pathological Q waves, and other
36 Advances in the Diagnosis of Coronary Atherosclerosis

non-specific ST segment and T wave changes. Second, any such changes may guide the choice
of the appropriate functional study. The presence of a left bundle branch block, for example,
essentially renders exercise stress electrocardiography uninterpretable and confounds
assessment of stress echocardiography or perfusion studies (La Canna et al, 1992).
Non-invasive tests such as exercise or pharmacological stress studies form an important
pathway in the diagnostic algorithm. The value of a functional study cannot be
overemphasized in patients with atypical or noncardiac chest pains. A negative functional
study, such a negative treadmill test to 12 minutes on the Bruce protocol, provides sufficient
prognostic information to negate any further investigations (Myers et al 2002; Marshall et al,
2010). The ischaemic burden as determined from imaging tests also provides pertinent
guidance as to the appropriateness of revascularization. A substudy (Shaw et al 2008) of the
COURAGE trial (Boden et al, 2007), which showed no benefit of PCI over optimized medical
therapy, did show mortality and MI benefit with revascularizing patients with >10%
ischaemic myocardial burden. A well executed functional study may also assist in
localizing the appropriate territory for revascularization, especially in the setting of multi-
vessel intermediate lesions.
Patients with positive stress tests, especially with high risk features (table three), suggestive
of significant ischaemic burden, should proceed to coronary angiography for risk
stratification with a view to possible percutaneous or surgical revascularization. Indeed, of
the indications of coronary angiography outlined in the ACC guideline for stable angina, a
positive stress test with high risk features carries the highest level of evidence (Level A)
compared to all other indications (level B to C).

Noninvasive test results predicting high risk for adverse outcome (> 3% annual mortality rate)
Severe resting left ventricular dysfunction (LVEF <35%)
High-risk treadmill score (score  -11)
Severe exercise induced left ventricular dysfunction (exercise LVEF <35%)
Stress-induced large perfusion defect (particularly if anterior)
Stress-induced moderate-size multiple perfusion defects
Large, fixed perfusion defect with left ventricular dilatation or increased lung uptake
Stress-induced moderate-size perfusion defect with left-ventricular dilatation or increased
lung uptake
Echocardiographic wall motion abnormality (involving >2 segments) developing at low
dose of dobutamine ( 10mg/kg/min) or at a low heart rate (<120bpm)
Stress echocardiographic evidence of extensive ischaemia

Table 3. High risk features in noninvasive tests


In some patients, however, it may be appropriate to consider coronary angiography as the
initial investigation to confirm coronary artery disease. Patients who are symptomatic
despite adequate or maximal antianginal therapy should proceed to coronary angiography
Indications for Coronary Angiography 37

with a view to revascularization for symptomatic benefit. As outlined in the ESC guideline
on myocardial revascularization (Wijns et al, 2010), patients deemed to have a high pretest
probability of coronary disease are advised against (IIIA and IIIB recommendations) non-
invasive tests but to proceed to coronary angiography (IA recommendation). Patients
intolerant of non-invasive testings, or with left ventricular dysfunction, could be considered
for coronary angiography. Cardiac transplant patients continue to form an important group
of the patients in whom coronary angiography is routinely performed for follow up for
transplant vasculopathy. It continues to be a class II recommendation for patients awaiting
organ transplant aged 40 years or older.

3.1.2 Coronary artery disease – stable pattern – known coronary artery disease
There is no role for surveillance routine coronary angiography in stable asymptomatic patients
with known CAD, previous coronary interventions or coronary bypass surgery, unless as
part of a research protocol. Patients who have documented coronary artery disease and
previous interventions or CABG, who have developed recurred or worsening symptoms,
coronary angiography is indicated and may be combined with percutaneous
revascularization. It is reasonable to proceed to coronary angiography directly in these
patients. Whilst functional non-invasive tests will still provide localizing data for inducible
ischaemia, intravascular testing modalities such as intravascular ultrasound, optical
coherence tomography and fractional flow reserve testing allow a robust evidence based
approach to guide any revascularization (Tonino et al 2009; Pijls et al 2007).

3.2 Coronary artery disease – unstable pattern


3.2.1 Acute coronary syndromes – with haemodynamic instability or rhythm
disturbance
Patients who presented with acute coronary syndromes with clinical instability (table 4)
should proceed to urgent angiography with a view to possible revascularization (Hochman
et al, 2006) either percutaneously or surgically if indicated.

Clinical instability in acute coronary syndromes

Cardiogenic shock
haemodynamic instability - severe hypotension on ischaemia
Recurrent/Persistent ischaemia and/or chest pain
Instability in cardiac rhythm e.g. major ventricular arrhythmia

Table 4. Clinical instability in acute coronary syndromes

3.2.2 Acute coronary syndromes – unstable angina and non ST elevation acute
coronary syndromes (non STEACS)
Non STEACS is the most frequent presentation of acute coronary syndromes and carries at
least as poor an outcome as STEACS (Chan et al 2009, Polonski et al, 2010). It is
recommended (ESC and ACC class I recommendation) that patients who presented with
non STEACS should be treated with an invasive strategy of coronary angiography. This is
especially for patients with evidence of ischaemia on ECG or biomarkers, the two most
38 Advances in the Diagnosis of Coronary Atherosclerosis

powerful predictors of adverse events, as a prelude to definitive treatment for unstable


coronary culprit lesions. Several meta-analyses including a Cochrane review (Hoenig et al,
2006; Fox et al, 2010; Mehta et al, 2005) of randomized controlled trials have concluded a
routine invasive strategy improved ischaemic endpoints - reducing recurrent ischaemia,
rehospitalisation and revascularization; and a more recent meta-analysis of three RCTs
(FRISC-II, ICTUS, RITA-3) in 2010 (Fox et al, 2010) showed the benefit of a routine invasive
strategy in cardiovascular death or non fatal MI was maintained at five years. Whilst the
meta-analysis was conclusive, one must note the heterogeneity in the trials included. The
most controversial trial, published in 2005, comparing a routine invasive vs. conservative
strategy, ICTUS (de Winter et al, 2005), was the only trial to show no difference in the
ischaemic end points between the two strategies. Five year follow up data from this study
continued to show no long-term benefit of an early invasive strategy in reducing death or
MI (Damman et al, 2010). Notably, 54% of the initially conservatively treated patients
received revascularization within one year. It is likely the ability of the ICTUS trial to
demonstrate difference in clinical outcomes has been significantly compromised by the cross
over rate (Cannon, 2004) from medical therapy to invasive treatment.
As a result of the heterogeneity between trials (inclusion criteria, definitions of MI, intensity
of pharmacological treatment and frequency of revascularization) and the confounding
results, this issue of routine invasive vs. conservative treatment strategy continues to spurn
passionate debate (Bittl & Maron, 2010; Thompson, 2010). The 2011 ACC guidelines
(Anderson et al, 2011) acknowledge that in selective patients without high-risk features who
have been stabilized, an initially conservative strategy may not be inappropriate, especially
those with serious co morbidities or contraindications to angiography and intervention who
have an elevated risk for clinical events, for example, patients with advanced malignancy
and a history of contrast allergy who declines invasive procedures. The decision to avoid
angiography in these patients may be made by considering the clinical evaluation of the
physician and patient preference. It is those patients enjoying satisfactory quality of life who
demonstrate the highest clinical risk profile that derive the most benefit and should receive
an early invasive strategy.
When an early invasive approach has been decided for a patient, the exact timing to
invasive coronary angiography is not very well established and subject to ongoing research.
This of course differs from the management in STEACS where urgent reperfusion is vital.
Various trials have attempted to answer the question of timing of coronary angiography in
NSTEACS. The risk of adverse cardiovascular events is time dependant. These events in the
unstable patient group are more common in the early phase after initial clinical
presentation. An early invasive treatment may prevent impending reinfarction or ischaemia,
but the counterargument is that an adequate pre procedural period of pharmacotherapy
may “prepare” the lesion better to minimize complications such as no reflow. The data is
not definitive. Small trials (Neumann et al, 2003), non-clinical primary endpoints
(Montalescot et al, 2009), possibly underpowered studies (Mehta et al, 2009) are all factors
that make definitive evidence based recommendations difficult.
Whilst the evidence differs between various trials, it is reasonable the low to intermediate
risk NSTEACS patients undergo coronary angiography within 72 hours. This is reflected in
the conservative arm of recent studies in whom a delayed approach is now considered a
median of 24 hours after the onset of symptoms compared to 72 hours 10 years ago. Based
on subgroup analysis on the TIMACS study in which patients with a GRACE score >140
Indications for Coronary Angiography 39

benefited most from an early invasive strategy, this group of high risk patients would
benefit from prompt (<24 hours) coronary angiography.

PatientsStudy design Median Primary Results Other findings


early vs. endpoint
Late
difference
(hrs)
TACTICS NSTEACS Angiography 22hrs vs. 6 month death, 15.9% vs.
TIMI 18 4-48 vs. 79 hrs MI, 19.4%
(2001) selective rehospitalization (p=0.025)
invasive for MI
ISAR- NSTEACS Angiography 2.4 hrs vs. 30 day death or 5.9% vs.
COOL < 6 hrs vs. > 86 hrs large MI 11.6%
(2003) 72 hrs (p=0.04)
TIMACS NSTEACS, Angiography 14 hrs vs. 6 month death, 9.6% vs. Refractory
(2009) <24 hrs of < 24 hrs vs. 50 hrs MI, stroke 11.3% ischaemia 3.3%
pain >36 hrs after (p=0.15) vs. 1.0%
randomization (p<0.001)
ABOARD NSTEACS Angiography 70 min Peak troponin I 2.1 vs. 20 death, MI,
(2009) “immediate” vs. 21 hrs during 1.7 urgent
vs. delayed hospitalization ng/mL revascularization
13.7% vs. 10.2%
p=0.31
Table 5. Timing of coronary angiography in NSTEACS

3.2.3 Acute coronary syndromes – ST elevation ACS


The cornerstone of management in ST elevation ACS is the timely restoration of flow in the
infarct related artery. The accepted strategies for reperfusion include primary PCI and
thrombolysis. If available within a timely fashion from an experienced team, urgent
coronary angiography and primary PCI offers better patient outcomes than thrombolysis in
STEACS. In addition, primary PCI is recommended in patients with cardiogenic shock or
contraindication to thrombolysis.
In patients with failed thrombolysis, or recurrent ischaemia after initially successful
thrombolysis, urgent coronary angiography should be considered, as shown in a meta-
analysis in 2007 (Wijeysundera et al, 2007). Rescue PCI is associated with significant risk
reduction in reinfarction and heart failure, as well as a trend towards mortality reduction. In
the REACT trial (Gerschlick et al, 2005), there was a significant reduction of the primary
composite end point of death, reinfarction, stroke or severe heart failure at six months for
rescue PCI, compared to both conservative treatment of repeat thrombolysis. Long term
data (Carver et al, 2009), median 4.4 years, from this trial reinforced the mortality reduction
of rescue PCI over both conservative treatment or repeat thrombolysis (RR 0.41, p=0.004; RR
0.43, p=0.006, respectively). Repeat thrombolysis is contraindicated. It is associated with
excessive bleeding and no benefit in reperfusion. In patients who presented to a non PCI-
capable centre, if transfer to a PCI-capable centre can be undertaken such that the delay
between first medical contact and balloon inflation is less than two hours, patient should be
40 Advances in the Diagnosis of Coronary Atherosclerosis

transferred to the PCI-capable centre. Established logistics and personnel arrangement must
be in place for this to take place.
In patients with successful thrombolysis, the timing of transfer to a PCI-capable facility and
the timing of angiography have been addressed in several contemporary trials and meta-
analyses. Several recent meta-analyses (Borgia et al, 2010; Desch et al, 2008; Savio et al, 2010)
which included studies such as CARESS-AMI (Di Mario et al, 2009), TRANSFER-AMI
(Cantor et al, 2010) and NORDISTEMI (Bøhmer et al, 2009), demonstrated that early transfer
to a PCI capable facility is associated with a significant reduction in the combined endpoint
of 30 day mortality, re-infarction, ischaemia. The benefit appears to be driven most by a
reduction of re-infarction and ischaemia. Current ESC and ACC guidelines recommend this
pharmacoinvasive strategy, or “drip and ship” strategy, the prompt transfer of
thrombolyzed patients to a PCI capable hospital within 24 hours.
The timing of coronary angiography deserves special mention. In stable patients
successfully thrombolyzed, having been transferred to a PCI capable facility, coronary
angiography and PCI should be deferred until 3 to 24 hours after thrombolysis. Whilst
immediate coronary angiography appears attractive a proposition, evidence from
“facilitated PCI” suggests otherwise. Facilitated PCI is a strategy of immediate angiography
and PCI after pharmacotherapy such as upstream full or half dose thrombolytic therapy,
with or without glycoprotein IIb/IIIa inhibitor. Several trials have addressed this question.
ASSENT-4 (ASSENT-4 investigators, 2006), assessing tenecteplase then PCI compared to
primary PCI, showed increased rates of ischaemic and bleeding complications with
facilitated PCI, and FINESSE (Ellis et al, 2008, reduced reteplace and abciximab then PCI vs.
abciximab then PCI vs. PCI) showed no difference in the mortality, ventricular function or
cardiogenic shock but excess bleeding. A meta-analysis (Keeley et al, 2006) of 18 trials over 3
decades showed no benefit of facilitated PCI over primary PCI. There is currently no data to
recommend the use of “facilitated” PCI. Pharmacoinvasive strategy is encouraged but
facilitated PCI is not.
Coronary angiography should be performed prior to definitive surgical treatment for
mechanical complications of any myocardial infarction (e.g. ventricular septal defect, acute
mitral regurgitation or ventricular aneurysm). There are some situation however where the
delays associated with angiography may outweigh the benefits of defining the coronary
anatomy. For example angiography should not delay emergent surgical treatment of VSD
and shock or aortic dissection.

3.2.4 Out of hospital cardiac arrest (OOHCA)


Patients who have survived an out of hospital cardiac arrest with return of spontaneous
circulation (ROSC) may or may not present with diagnostic ischaemic ECG changes. In
patients with definitive changes such as ST elevation, they should be treated as outlined
above with an immediate invasive strategy. In haemodynamically and rhythmically stable
patients after cardiac arrest, the appropriate timing (immediate vs. deferred) of coronary
angiography is the subject of particular interest. Up to 80% of OOHCA patients have had an
acute coronary syndrome as the precipitant (Pell et al, 2003) and ECG changes are not
necessarily predictive (Aurore et al, 2011). Given this finding, it was not surprising some
studies (Dumas et al, 2010; Reynolds et al, 2009; Spaulding et al, 1997; Sunde et al, 2007; Stub
et al 2010) have shown an improved outcome, including cardiac ejection fraction,
Indications for Coronary Angiography 41

Patients Study design Primary Results Hrs to PCI Other


endpoints in transfer findings
group
CARESS-in- 75 y.o. or Immediate 30 day RR 0.40 <3
AMI (2008) younger transfer for death, (p=0.004) (median 2.2)
STEACS, ½ angiography reinfarction,
reteplace ±PCI vs. refractory
with standard ischaemia
abciximab, treatment
heparin, (with transfer
aspirin for rescue if
within 12 needed)
hours of
pain
TRANSFER- STEACS, Immediate 30 day RR 0.64 <6
AMI (2009) transfer for
tenecteplase death, (p=0.004) (median 3.9)
angiography±
within 12 reinfarction,
hours ofPCI within 6 recurrent
pain hours vs. ischaemia,
standard- new CCF,
treatment cardiogenic
(deferred > shock
24 hrs)
NORDISTEMI 75 y.o. or Immediate 12 month RR 0.72 As soon as 30 day
(2010) younger, angiography death, (NS) possible death,
STEACS, ±PCI vs. reinfarction, (median 2.7) reinfarction,
teneteplase Deferred stroke, new stroke or
<6 hr of ischaemia new
pain, ischaemia
tenecteplase (RR 0.49
p=0.03)

Table 6. Timing of transfer of STEACS patients to a PCI capable facility


neurological recovery and survival, with this strategy. The largest study of this patient
group is the PROCAT registry (Dumas et al, 2010) from Paris suggested that immediate
angiography and successful PCI improved survival (odds ratio 2.06, p=0.013). Given the
evidence leading centers have adoped a strategy of immediate coronary angiography and
revascularisation as indicated after an OOHCA with ROSC. Indeed, this is the
recommendation endorsed by the International Liaison Committee on Resuscitation
(ILCOR, 2010) but the level of evidence is relatively low. Coma or significant neurological
compromise are common in this patient group and these should not preclude consideration
of angiography with full neurological recovery in initially comatose STEACS patients found
in various registries (Garot et al, 2007; Hosmane et al, 2009). Further, the induction of
42 Advances in the Diagnosis of Coronary Atherosclerosis

hypothermia (Bernard et al, 2002; Wolfrum et al, 2008) should be incorporated as part of the
treatment strategy for those undergoing immediate angiography.

3.3 Special considerations


3.3.1 Congestive cardiac failure
Invasive coronary angiography is useful in the investigation of left ventricular systolic
dysfunction for an ischemic aetiology, especially in the presence of suggestive features such
as angina, remote history of severe angina or even silent infarction, ischaemia on surface
electrocardiogram, or regional wall motion abnormalities on cardiac imaging. Since the 1999
ACC guideline, the advance of non-invasive computed tomography coronary angiography
(Andreini et al, 2009), with its high sensitivity and negative predictive value, may justify its
use for this particular indication rather than invasive coronary angiography. The negative
predictive value of a normal scan has been shown to approach 100%.

3.3.2 Preoperative assessment for noncardiac surgery


The premise of preoperative cardiac, in particular coronary, assessment for noncardiac
surgery is to prevent peri operative cardiac adverse events and mortality. Surgery places
significant physiological stresses that may cause cardiac ischaemia from stenosed epicardial
arteries as well as inflammation and activation of the coagulation cascade possibly
precipitating acute coronary syndromes.
As a general rule, the patient should be managed as would be in a non-surgical setting and
testing of any type should only be undertaken if it is likely to change peri operative
management. As such, in any emergency life saving operations, it is accepted any cardiac
investigations should not be performed.
Coronary angiography has a limited role in the routine management of patients planned for
non-cardiac surgery. A comprehensive discussion on the preoperative assessment of
noncardiac surgery is beyond the scope of this chapter but the key determinants are: patient
symptoms and clinical risk profile, surgical risk estimate (table 7) (Boersma et al, 2005), and
the patient’s functional capacity.

Surgical risk estimate (adapted from Boersma et al, 2005) – risk of MI and cardiac death at
30 days
Low risk <1% Intermediate risk 1-5% High risk >5%
Breast Abdominal Aortic and major
vascular surgery
Dental Carotid Peripheral vascular
surgery
Endocrine Peripheral arterial angioplasty
Eye Endovascular aneurysm repair
Reconstructive Head and neck surgery
Orthopaedic – minor e.g. Neurological/major
knee orthopaedic
Urologic – minor Pulmonary/renal/liver
transplant
Urologic – major
Table 7. Risk of adverse cardiac events in different surgical scenarios
Indications for Coronary Angiography 43

Patients who have stable angina or are asymptomatic can generally proceed with surgery.
Patients who have worsening symptoms of ischaemia probably deserve further assessment,
namely in the form of functional studies, prior to elective surgery. The same indications for
coronary angiography and/or intervention should apply here as would be in the non-
surgical setting. The cardiac risks of the particular operation should be assessed. It is a class
I recommendation (Poldermans et al, 2009) that pre-operative coronary angiography be
performed in acute coronary syndromes as well as in patients with angina refractory to
medical therapy. Pre-operative angiography is not recommended in cardiac-stable patients
undergoing low-risk and even intermediate risk surgery.
Evidence of improved outcome with revascularization prior to surgery however is lacking.
There is little data pertaining to the benefit of revascularization in patients undergoing non
cardiac vascular surgery, a subset of surgical patients known for high cardiac co-morbidity.
One study (McFalls et al, 2004 - Coronary Artery Revascularization Prophylaxis CARP
study) on stable CAD patients with preserved LV failed to demonstrate improved outcome
with prophylactic revascularization compared to optimized medical therapy. Another study
(Poldermans et al, 2007 - DECREASE-V pilot study), assessed the same concept on complex
CAD with impaired LV systolic function, and reached the same conclusion. This was despite
75% of these cases having three vessel or left main coronary artery disease. On the currently
limited available data, prophylactic revascularization cannot be recommended.

3.3.3 Non invasive computed tomography coronary angiography (CTCA)


Over the past decade the technological advances in CTCA have meant that the technology is
emerging as an important investigation in a range of clinical scenarios. Aside from its non-
invasive nature, CTCA provides information about both vessel lumen and important
information on the composition of plaque and the vessel wall as well as other cardiac
structures. However, it is important to note the spatial and temporal resolutions remain
inferior to invasive coronary angiography with the current technology. Various multicentre
studies (Budoff et al, 2008; Miller et al, 2008; Mowatt et al, 2008) on 64 slice CTCA have been
published and confirmed its inferiority to invasive coronary angiography particularly in its
specificity and the limitation on small vessel definition (table 8). The evidence for its clinical
application continues to evolve.

Study Sensitivity Specificity Sensitivity Specificity Negative Positive


author per vessel per vessel per patient per predictive predictive
patient value value
(per atient) (per patient)
Budoff et al 84% 90% 95% 83% 99% 64%
2008
Meijboom 95% 77% 99% 64% 97% 86%
et al 2008
Miller et al 75% 93% 85% 90% 83% 91%
2008

Table 8. Meta-analyses of accuracy 64 slice CTCA compared to coronary angiography


44 Advances in the Diagnosis of Coronary Atherosclerosis

Guidelines on the appropriateness of CTCA (Taylor et al, 2010) published in 2010 attempted
to address the use of this rapidly evolving technology. The indications chosen mirrored that
of invasive coronary angiography. In view of its high sensitivity, CTCA is particularly
suited to low to intermediate risk patients in the setting of stable symptomatic patients, low
risk acute coronary syndromes and heart failure patients. Patients with normal CTCA
findings convey a very low risk of future cardiac events and cardiac mortality (Hulten et al,
2011). Patients at high risk of coronary artery disease are advised against CTCA as this
represents hindrance to timely diagnosis and management of CAD with the additional risk
of contrast and radiation. It has no role in the preoperative assessment of non-cardiac
surgery. It is deemed appropriate in the assessment of graft patency for symptomatic
patients with previous CABG. To reflect the emerging role of left main stenting, CTCA has
been deemed appropriate as a means for routine follow up screening of left main stent(s) in
asymptomatic patients.
Unless the limitations of CTCA can be sufficiently improved upon, it is likely CTCA will
continue to be utilized in a complementary role to invasive coronary angiography in the
assessment of CAD. Of course the major advantage of angiography remains the ability to
proceed on to coronary intervention as part of the imaging procedure.

3.4 Relative and absolute contraindications


The only absolute contraindication is if a patient refuses the test fully understanding the risk
and benefit of his/her decision, refuses any treatment for CAD regardless of the result of the
coronary angiography, or if other medical co-morbidities rendering imaging of the
coronaries and revascularization inappropriate or impossible. Subjecting the patient to even
the small risks of a coronary angiogram with no foreseeable gain cannot be justified.
Relative contraindications include situations which increase the likelihood of complications,
e.g. bleeding diathesis, coagulopathy (iatrogenic i.e. warfarin or pathological), renal
impairment, anaemia, contrast allergy, active infection and aortic valve vegetation. If these
relative contraindications are reversible the patient should be appropriately prepared and
coronary angiography deferred.

4. Valvular heart disease and structural heart disease intervention


The assessment of the coronary anatomy prior to cardiac valvular surgery is routine
although not necessarily evidence driven. Whilst surgery for valvular heart disease has been
performed for many years, percutaneous intervention for structural or valvular heart
disease has been generating significant interest over the recent years. Transcatheter aortic
valve implants (TAVI) have already demonstrated significant mortality benefit in patients
deemed unsuitable for surgery compared to medical therapy, in the first RCT on TAVI
(PARTNERS trial) (Leon et al, 2010). Several investigational percutaneous treatment options
also exist for mitral regurgitation. Percutaneous coronary revascularization has been a
prerequisite for the TAVI procedures and as such all patients underwent coronary
angiography. The incidence of coronary disease in this patient group is high, up to 70% the
PARTNERS trial. Coronary angiography is a pivotal part in the assessment for TAVI
suitability. The increasing percutaneous treatment options will likely generate in the future
specific college endorsed guideline to incorporate the role of coronary angiography in pre
structural heart intervention..
Indications for Coronary Angiography 45

4.1 Pre valvular surgery


It is routine to perform coronary angiography prior to valve surgery although there is no
evidence to guide such practice (Bonow et al, 2006; Vahanian et al, 2007). This is especially
logical in older patients with cardiac risk factors, or suggestion of CAD from symptoms,
ECG, cardiac imaging or non-invasive function studies. The indication is less clear in
younger patients, and special caution must be exercised in aortic valve surgery for infective
endocarditis given the risk for vegetation embolization. No definitive data are available to
define the clinical benefit of revascularization in the setting of corrective valve surgery.
Whilst it is reasonable to expect benefit in prognostic disease, the same generalization may
not apply to single vessel non prognostic lesions which would otherwise not necessarily be
revascularized in a non-surgical setting. The role of CTCA is yet to be defined but its
application here may reasonably mimic that outlined previously, in a low to intermediate
risk group of patients undergoing valvular surgery (Meijboom et al, 2006).

4.1.1 Pre percutaneous valve implant


This is a developing area in which the role of coronary angiography appears crucial.
Coronary angiography and if necessary percutaneous revascularization were a requisite for
all TAVI trials so as to allow for direct comparison with the CABG + AVR subgroup. It
seems nonetheless reasonable for this group of high risk patients, often with symptoms
indistinguishable from ischaemia, to undergo coronary angiography. In addition, the left
heart catheterization is pivotal for necessary measurement of aortic dimensions, angulation,
coronary ostia position and origin, and assessment of vascular access. It should be noted the
indication of revascularization in patients undergoing TAVI stems from its equivalent in
surgical treatment, the exact timing and completeness of revascularization are yet to be
defined.
The more difficult question relates to the need for coronary angiography in patients who
may not otherwise meet the indications for coronary angiography, e.g. pulmonary valve
implant in a young congenital heart disease patient, mitral valve clip in severe functional
mitral regurgitation. These patients are managed the patient in a similar fashion to the
conventional indications for coronary angiography.

4.1.2 Other structural cardiac interventions


There is limited evidence to support routine coronary angiography in structural cardiac
interventions such as patent foramen ovale and atrial septal defect closure, coarctation
stenting, patent ductus arteriosus interventions. It is often performed however based on the
individual patient’s clinical risk profile. The discovery of prognostically significant coronary
artery disease not amenable to percutaneous coronary interventions may sway treatment
towards a complete surgical treatment option. Coronary angiography does not increase the
invasive nature of these procedures and add little incremental risk.

5. Case vignettes of the use of coronary angiography in the contemporary


era
Case One – atrial septal defect with multi-vessel coronary artery disease
A 73 year old female presented with NYHA (New York Heart Association) class II
exertional dyspnoea. Her cardiac risk factors included hypertension and hyperlipidaemia. A
46 Advances in the Diagnosis of Coronary Atherosclerosis

transthoracic echocardiogram demonstrated two small secundum atrial septal defects


(ASD), with moderate right ventricular dilatation and normal systolic function.
Percutaneous ASD closure was attempted. Coronary angiography was performed,
especially given the uncharacteristically late presentation of her ASD, as well as her cardiac
risk factors. This demonstrated prognostically significant triple vessel disease (Figure 1a, 1b,
2c), with an osital 90% left circumflex artery stenosis, 90% mid left anterior descending
artery bifurcation stenosis and a 90% distal right coronary artery stenosis. Given the
unfavourable percutaneous prospect for the coronary lesions, she was referred for surgical
complete revascularization and ASD closure. She was discharged uneventfully one week
after surgery. Coronary angiography should be considered prior to structural cardiac
interventions in patients with cardiac risk factors and may alter management.

Fig. 1a. Left anterior artery stenoses


Indications for Coronary Angiography 47

Fig. 1b. Ostial left circumflex stenosis

Fig. 1c. Posterolateral branch stenosis


48 Advances in the Diagnosis of Coronary Atherosclerosis

Case Two – post TAVI angiogram


An 84 year old male represented with angina two years after a successful transcatheter
aortic valve implant (TAVI) with a bioprosthetic CoreValve®. His NYHA symptomatology
improved from Class IV to Class I. His coronary angiogram and graft study prior to his
TAVI showed patent grafts (LIMA-LADD1-LAD; SVG-OM1; SVG-PDA) but severe native
disease including a severely diseased grafted PDA with no prospect for surgical or
percutaneous revascularization. A coronary angiogram and graft study was performed
through his CoreValve® to assess if his underlying disease had progressed (Figure 2a).

Fig. 2a. Angiography with a 5 French Judkins Left 5 catheter through a CoreValve ®
bioprosthesis
Indications for Coronary Angiography 49

This demonstrated patent grafts as previously noted but progression in his PDA disease
(Figure 2b). Percutaneous, or surgical, treatment remained an unfavourable prospect and he
was successfully treated with intensified medical therapy with CCS (Canadian
Cardiovascular Society) I angina. Coronary angiography can be safely performed through
CoreValve struts but care must be exercised with the choice and manipulation of catheters.

Fig. 2b. Severe native posterior descending artery disease not amenable to percutaneous
treatment
Case Three – percutaneous treatment of left main stenosis and aortic stenosis
A 76 year old male with severe aortic stenosis was referred for possible percutaneous
treatment due to his high surgical risk. Assessment for TAVI entailed a coronary angiogram,
aortogram and an iliofemoral angiogram (Figure 3a). Coronary angiography revealed a
significant left main stenosis (Figure 3b) which on intravascular ultrasound measured 5.8
mm2. A multidisciplinary heart team assessed the data and concluded surgical treatment to
be prohibitively high risk. The patient then underwent left main stenting with a drug eluting
stent under IVUS guidance. Satisfactory stent expansion was attained with a final diameter
of 4.5mm (Figure 3c). The patient then proceeded to a successful TAVI (CoreValve®)
implant and continued to enjoy excellent symptom relief at six months.
50 Advances in the Diagnosis of Coronary Atherosclerosis

Coronary angiography, and arterial (aortic, iliac, and femoral) assessment, form an integral
part of TAVI assessment. Elderly patients with severe aortic stenosis often have severe co-
existing coronary artery disease as well as peripheral vascular disease as demonstrated by
this case. Optimizations of the coronary status, as well as an appreciation of the peripheral
vasculature, are absolutely paramount to a successful TAVI implant and to avoid vascular
complications.

Fig. 3a. Tortuous aorta and iliac arteries, important information for TAVI transfemoral
access.
Indications for Coronary Angiography 51

Fig. 3b. Mid shaft left main stenosis


52 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 3c. Drug eluting stent deployment in left main guided by IVUS
Case Four – anomalous coronary artery
A 63 year old male underwent coronary angiography after being successfully thrombolyzed
for an inferior STEACS, 24 hours after his original pain onset. Coronary angiography via his
right femoral artery showed a normal left coronary system but the origin of the right
coronary artery (RCA) was not in the usual sinus. An Amplatz left catheter 2 (AL2)
eventually revealed (Figure 4a) an anomalous RCA from the opposite sinus (ACAOS). A
95% stenosis in the mid RCA was found and after trying various catheters, an EBU (extra
back up) 3.5 guiding catheter (Medtronic ®), traditionally a left coronary catheter, made it
possible for a successful intervention and the placement of a coronary stent (Figure 4b and
4c). A CTCA (Figure 4d) was performed to ascertain the course of the RCA and confirmed
ACAOS. Percutaneous coronary intervention in anomalous coronary arteries required
innovative use of guiding catheters.
Indications for Coronary Angiography 53

Fig. 4a. Right coronary artery injection with a 5Fr AL2 catheter showing an anomalous right
coronary origin with impossible selective engagement
54 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 4b. A 6Fr EBU catheter used to engage the anomalous origin. Traditional right coronary
catheters would not have been useful.
Indications for Coronary Angiography 55

Fig. 4c. Successful placement of a drug eluting stent in the mid RCA with no residual
stenosis.
56 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 4d. CTCA 3D reconstruction shows the origin and course of the anomalous right
coronary artery clearly and defines its ante-aortic course.
Case Five – CTCA in graft assessment
A 69 year old male developed atypical chest pain 12 months after his coronary bypass graft
(LIMA-LAD; RA-ramus; SVG-OM1). During the initial angiographic assessment he
underwent IVUS of his left main and left anterior descending artery indeterminate lesions.
IVUS showed his LM to be 5.44mm2 and LAD 3.96mm2. These were thought be suggest
significant stenoses. To investigate his current atypical chest pain an MPS (Figure 5a) was
performed which showed no inducible ischaemia or symptoms. A CTCA (Figure 5b) was
performed which however suggested possible occluded LIMA graft. A coronary angiogram
and graft study (Figure 5c and 5d) confirmed this finding that the LIMA graft had become
atretic and normal coronary flow seen in the LAD. His other grafts were patent as was
shown on the CTCA. This case demonstrated the value of CTCA in identifying graft patency
and position, often facilitating the location of the graft anastomosis at the aorta. The atypical
symptoms, patent coronary arteries and grafts, correlated well with his negative functional
study. The LIMA-LAD graft probably underwent atresia as a result of no physiological
implication to his intermediate LM and LAD lesions even though the diameters measured
fulfilled conventional criteria for significant lesions.
Indications for Coronary Angiography 57

Fig. 5a. Myoperfusion study with no significant inducible ischaemia.


58 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 5b. Position of two aortic grafts demonstrated clearly on CTCA. No LIMA flow
demonstrated.
Indications for Coronary Angiography 59

Fig. 5c. LAD artery with no obvious proximal stenosis and normal coronary flow.
60 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 5d. Small atretic LIMA.


Case Six – balanced ischaemia on MPS in a high risk patient
A 67 year old male with 20 years of diabetes mellitus II and 40 pack year history of smoking
presented with six months of worsening exertional dyspnoea and chest pain. A
myoperfusion study performed at a satisfactory level of workload demonstrated a small
area of mildly reversible ischaemia in the inferolateral area. Given the abnormal findings,
the patient proceeded to coronary angiography. This revealed severe distal left main artery
stenosis, with a subtotally occluded left circumflex and a proximal severe RCA stenosis. Left
ventricular function was normal. The extent and severity of disease was of prognostic
significance. The patient underwent uneventful coronary bypass grafting and was
discharged from the hospital one week after. This patient exhibited a high pretest
probability based on the clinical profile. Whilst the myoperfusion study was suggestive, it
underestimated the extent of disease likely due to the balanced ischaemia from the RCA and
LM stenoses. Complex distal left main stenosis, especially bifurcation stenosis, is best
treated with surgical revascularization.
Indications for Coronary Angiography 61

Fig. 6a. Distal left main eccentric stenosis and ostial circumflex stenosis.
62 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 6b. Proximal right coronary artery stenosis


Indications for Coronary Angiography 63

6. Conclusion
Coronary angiography remains the most accurate diagnostic modality for coronary artery
disease. It provides a definitive assessment of coronary artery lumen and provides an
avenue to percutaneous intervention. It is pivotal in the treatment on unstable or acute
coronary syndromes and life saving in cardiogenic shock and STEACSs. Indications for
invasive coronary angiography have changed incrementally over the decade since
ACC/AHA guidelines on the topic were published. The advent of noninvasive CTCA has
changed the landscape for the diagnosis of CAD but CTCAs seem destined only for the low
to intermediate risk patients. Coronary angiography also serves a pivotal role in the
burgeoning field of percutaneous structural cardiac interventions, especially percutaneous
valve implants.

7. References
Andreini D, Pontone G, Bartorelli AL, Agostoni P, Mushtaq S, Bertella E, Trabattoni D,
Cattadori G, Cortinovis S, Annoni A, et al. (2009). Sixty-Four-Slice Multidetector
Computed Tomography: An Accurate Imaging Modality for the Evaluation of
Coronary Arteries in Dilated Cardiomyopathy of Unknown Etiology Circ
Cardiovasc Imaging 2(3): 199 - 205.
Anderson JL, Adams CD, et al. (2011). 2011 ACCF/AHA Focused Update Incorporated into
the ACC/AHA 2077 Guidelines for the management of patients with unstable
angina/Non-ST Elevation myocardial infarction, A report of the American College
of Cardiology Foundation/American Heart Association Task Force on Practice
Guidelines, Circulation. 2011; 123 [Epub ahead of print]
ASSENT-4 investigators, (2006). Primary versus tenecteplase-facilitated percutaneous
coronary intervention in patients with ST-segment elevation acute myocardial
infarction (ASSENT-4 PCI): randomised trial. Lancet 367:569–578.
Aurore A, Jabre P, Liot P, Margenet A, Lecarpentier E, Combes X. (2011). Predictive factors
for positive coronary angiography in out-of-hospital cardiac arrest patients. Eur J
Emerg Med. 18(2):73-6.
Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Gutteridge G, et al. (2002).
Treatment of comatose survivors of out-of-hospital cardiac arrest with induced
hypothermia. N Engl J Med 346: 557 – 563
Bittl JA, Maron DJ. (2010). Having It Both Ways J. Am. Coll. Cardiol., 55(9): 865 - 866.
Boden WE, O'Rourke RA, Teo KK, et al. (2007). Optimal medical therapy with or without
PCI for stable coronary disease. N Engl J Med 356:1503-1516
Boersma E, Kertai MD, Schouten O, Bax JJ, Noordzij P, Steyerberg EW, Schinkel AF, van
Santen M, Simoons ML, Thomson IR, Klein J, van Urk H, Poldermans D. (2005).
Perioperative cardiovascular mortality in noncardiac surgery: validation of the Lee
cardiac risk index. Am J Med 118:1134–1141.
Bøhmer E, Hoffmann P, Abdelnoor M, Arnesen H, Halvorsen S. (2010). Efficacy and safety
of immediate angioplasty versus ischemia-guided management after thrombolysis
in acute myocardial infarction in areas with very long transfer distances: results of
64 Advances in the Diagnosis of Coronary Atherosclerosis

the NORDISTEMI (NORwegian study on DIstrict treatment of ST-Elevation


Myocardial Infarction) J Am Coll Cardiol 55:102-110.
Bonow RO, Carabello BA, Kanu C, de Leon AC Jr, Faxon DP, Freed MD, Gaasch WH, Lytle
BW, Nishimura RA, O'Gara PT, O'Rourke RA, Otto CM, Shah PM, Shanewise JS,
Smith SC Jr, Jacobs AK, Adams CD, Anderson JL, Antman EM, Faxon DP, Fuster V,
Halperin JL, Hiratzka LF, Hunt SA, Lytle BW, Nishimura R, Page RL, Riegel B.
(2006). ACC/AHA 2006 guidelines for the management of patients with valvular
heart disease: a report of the American College of Cardiology/American Heart
Association Task Force on Practice Guidelines (writing Committee to Revise the
1998 guidelines for the management of patients with valvular heart disease)
developed in collaboration with the Society of Cardiovascular Anesthesiologists
endorsed by the Society for Cardiovascular Angiography and Interventions and the
Society of Thoracic Surgeons. J Am Coll Cardiol. 48: e1–e148.
Borgia, F, Goodman SC, Halvorsen S, Cantor WJ, Piscione F, Le May MR, Fernandez-Aviles
F, Sanchez PL, Dimopoulos K, Scheller B, Armstrong PW and Di Mario C. (2010).
Early routine Percutaneous coronary intervention after fibrinolysis vs. standard
therapy in ST-segment elevation myocardial infarction: a meta-analysis, Eur Heart J
31 (17):2156-2169
Budoff MJ, Dowe D, Jollis JG, et al. (2008). Diagnostic performance of 64-multidetector-row
coronary computed tomographic angiography for evaluation of coronary artery
stenosis in individuals without known coronary artery disease. J Am Coll Cardiol
52:1724–32.
Cannon CP. (2004). Revascularisation for everyone? Eur Heart J 25:1471–1472
Cantor WJ, Fitchett D, Borgundvaag B, Ducas J, Heffernan M, Cohen EA, Morrison LJ,
Langer A, Dzavik V, Mehta SR, Lazzam C, Schwartz B, Casanova A, Goodman SG.
(2009). Routine early angioplasty after fibrinolysis for acute myocardial infarction.
N Engl J Med 360:2705–2718.
Carver A, Rafelt S, Gershlick AH, et al. REACT Investigators. (2009). Longer-term follow-up
of patients recruited to the REACT (Rescue Angioplasty Versus Conservative
Treatment or Repeat Thrombolysis) trial J Am Coll Cardiol 54:118-126.
Chan MY, Sun JL, Newby LK, Shaw LK, Lin M, Peterson ED, Califf RM, Kong DF, Roe MT.
(2009). Long-term mortality of patients undergoing cardiac catheterization for ST-
elevation and non-ST elevation myocardial infarction, Circulation. 19(24):3110-7
Damman P, Hirsch A, Windhausen F, Tijssen JGP, de Winter RJ, ICTUS Investigators.
(2010). 5-year clinical outcomes in the ICTUS (Invasive versus Conservative
Treatment in Unstable coronary Syndromes) trial: a randomized comparison of an
early invasive versus selective invasive management in patients with non–ST-
segment elevation acute coronary syndrome J Am Coll Cardiol 55:858-864.
de Winter RJ, Windhausen F, Cornel JH, et al (2005). Invasive versus Conservative
Treatment in Unstable Coronary Syndromes (ICTUS) Investigators. Early invasive
versus selectively invasive management for acute coronary syndromes. N Engl J
Med 353: 1095-1104.
Desch S, Eitel I, Rahimi K, de Waha S, Schuler G, Thiele H, (2010). Timing of invasive
treatment after fibrinolysis in ST elevation myocardial infarction – a meta-analysis
Indications for Coronary Angiography 65

of immediate or early routine versus deferred or ischaemia-guided randomized


controlled trials, Heart, 96(21):1695-1702
Di Mario C, Dudek D, Piscione F, Mielecki W, Savonitto S, Murena E, Dimopoulos K,
Manari A, Gaspardone A, Ochala A, Zmudka K, Bolognese L, Steg PG, Flather M.
(2008). Immediate angioplasty versus standard therapy with rescue angioplasty
after thrombolysis in the Combined Abciximab Reteplase Stent Study in Acute
Myocardial Infarction (CARESS-in-AMI): an open, prospective, randomised,
multicentre trial. Lancet 371:559–568
D'Souza SP, Mamas MA, Fraser DG, Fath-Ordoubadi F. (2010). Routine early coronary
angioplasty versus ischaemia-guided angioplasty after thrombolysis in acute ST-
elevation myocardial infarction: a meta-analysis Eur Heart J first published online
October 28, 2010
Dumas F, Cariou A, Manzo-Silberman S, et al. (2010) Immediate percutaneous coronary
intervention is associated with better survival after out-of-hospital cardiac arrest:
insights from the PROCAT (Parisian Region Out of hospital Cardiac ArresT)
registry. Circ Cardiovasc Interv 3:200–7
Ellis SG, Tendera M, de Belder MA, van Boven AJ, Widimsky P, Janssens L, Andersen HR,
Betriu A, Savonitto S, Adamus J, Peruga JZ, Kosmider M, Katz O, Neunteufl T,
Jorgova J, Dorobantu M, Grinfeld L, Armstrong P, Brodie BR, Herrmann HC,
Montalescot G, Neumann FJ, Effron MB, Barnathan ES, Topol EJ. (2008). Facilitated
PCI in patients with ST-elevation myocardial infarction. N Engl J Med 358:2205–
2217.
Fernandez-Aviles F, Alonso JJ, Castro-Beiras A, Vazquez N, Blanco J, Alonso-Briales J,
Lopez-Mesa J, Fernandez-Vazquez F, Calvo I, Martinez-Elbal L, San Roman JA,
Ramos B. (2004). Routine invasive strategy within 24 hours of thrombolysis
versus ischaemia-guided conservative approach for acute myocardial infarction
with ST-segment elevation (GRACIA-1): a randomised controlled trial. Lancet
364:1045–1053
Fox KA, Clayton TC, Damman P, Pocock SJ, de Winter RJ, Tijssen JG, Lagerqvist B,Wallentin
L. (2010). Long-term outcome of a routine versus selective invasive strategy in
patients with non-ST-segment elevation acute coronary syndrome a meta-analysis
of individual patient data. J Am Coll Cardiol 55:2435–2445.
Garot P, Lefevre T, Eltchaninoff H, Morice MC, Tamion F, Abry B, Lesault PF, Le Tarnec JY,
Pouges C, Margenet A, Monchi M, Laurent I, Dumas P, Garot J, Louvard Y. (2007).
Six-month outcome of emergency percutaneous coronary intervention in
resuscitated patients after cardiac arrest complicating ST-elevation myocardial
infarction. Circulation. 115: 1354–1362.
Gershlick AH, Stephens-Lloyd A, Hughes S, et al. (2005). Rescue angioplasty after failed
thrombolytic therapy for acute myocardial infarction. N Engl J Med. 353:2758–
68
Hoenig MR, Doust JA, Aroney CN, Scott IA. (2006). Early invasive versus conservative
strategies for unstable angina & non-ST-elevation myocardial infarction in the stent
era. Cochrane Database Syst Rev 3: CD004815.
66 Advances in the Diagnosis of Coronary Atherosclerosis

Hosmane VR, Mustafa NG, Reddy VK, et al. (2009). Survival and neurologic recovery in
patients with ST-segment elevation myocardial infarction resuscitated from cardiac
arrest. J Am Coll Cardiol 53:409–15.
Hulten EA, Carbonaro S, Petrillo SP, Mitchell JD, Villines TC. (2011). Prognostic value of
cardiac computed tomography, A systematic review and meta-analysis, J. Am. Coll.
Cardiol 57:1237-1247.
ILCOR, Part 9: Acute coronary syndromes 2010 International Consensus on
Cardiopulmonary Resuscitation and Emergency Cardiovascular Care Science with
Treatment Recommendations_Nolan, JP, Hazinski MF, et al, Part 1: Executive
summary 2010 International Consensus on Cardiopulmonary Resuscitation and
Emergency Cardiovascular Care Science With Treatment Recommendations
Resuscitation 81S (2010) e175–e212Resuscitation 81S (2010) e1–e25
Keeley EC, Boura JA, Grines CL. (2006). Comparison of primary and facilitated
percutaneous coronary interventions for ST-elevation myocardial infarction:
quantitative review of randomised trials Lancet 367:579-588
Leon MB, Smith CR, Mack M, et al. (2010). Transcatheter aortic-valve implantation for aortic
stenosis in patients who cannot undergo surgery. N Engl J Med 2010;
DOI:10.1056/NEJMoa1008232
McFalls EO, Ward HB, Moritz TE, Goldman S, Krupski WC, Littooy F, Pierpont G, Santilli S,
Rapp J, Hattler B, Shunk K, Jaenicke C, Thottapurathu L, Ellis N, Reda DJ,
Henderson WG. (2004). Coronary-artery revascularization before elective major
vascular surgery. N Engl J Med 351:2795 – 2804.
Mehta SR, Cannon CP, Fox KA, Wallentin L, Boden WE, Spacek R, Widimsky P,
McCullough PA, Hunt D, Braunwald E, Yusuf S. (2005). Routine vs selective
invasive strategies in patients with acute coronary syndromes: a collaborative
meta-analysis of randomized trials. JAMA 293:2908–2917.
Mehta SR, Granger CB, Boden WE, et al. (2009). Early versus delayed invasive intervention
in acute coronary syndromes. N Engl J Med. 360: 2165–75.
Meijboom WB, Meijs MF, Schuijf JD, et al. (2008) Diagnostic accuracy of 64-slice computed
tomography coronary angiography: a prospective, multicenter, multivendor study.
J Am Coll Cardiol 52:2135-44
Meijboom WB, Mollet NR, Van Mieghem CAG, Kluin J, Weustink AC, Pugliese F,
Vourvouri E, Cademartiri F, Bogers AJJC, Krestin GP, et al. (2006). Pre-Operative
Computed Tomography Coronary Angiography to Detect Significant Coronary
Artery Disease in Patients Referred for Cardiac Valve Surgery J. Am. Coll. Cardiol.,
48(8): 1658 - 1665.
Miller JM, Rochitte CE, Dewey M, et al. (2008). Diagnostic performance of coronary
angiography by 64-row CT. N Engl J Med 359:2324–36.
Montalescot G, Cayla G, Collet JP, et al. (2009). Immediate vs delayed intervention for acute
coronary syndromes: a randomized clinical trial. JAMA. 302:947–54.
Mowatt G, Cook JA, Hillis GS, et al. (2008). 64-Slice computed tomography angiography in
the diagnosis and assessment of coronary artery disease: systematic review and
meta-analysis. Heart 94:1386–93.
Indications for Coronary Angiography 67

Neumann FJ, Kastrati A, Pogatsa-Murray G, et al. (2003). Evaluation of prolonged


antithrombotic pretreatment (“cooling-off” strategy) before intervention in patients
with unstable coronary syndromes: a randomized controlled trial. JAMA. 290:1593–
9.
Pell JP, Sirel JM, Marsden AK, Ford I, Walker NL, Cobbe SM. (2003). Presentation,
management, and outcome of out of hospital cardiopulmonary arrest: comparison
by underlying aetiology. Heart 89:839—42
Poldermans D, Bax JJ, Boersma E, De Hert S, Eeckhout E, Fowkes G, Gorenek B, Hennerici
MG, Iung B, Kelm M, Kjeldsen KP, Kristensen SD, Lopez-Sendon J, Pelosi P,
Philippe F, Pierard L, Ponikowski P, Schmid JP, Sellevold OF, Sicari R, Van den
Berghe G, Vermassen F, Hoeks SE, Vanhorebeek I. (2009). Guidelines for
preoperative cardiac risk assessment and perioperative cardiac management in
noncardiac surgery: the Task Force for Preoperative Cardiac Risk Assessment and
Perioperative Cardiac Management in Non-cardiac Surgery of the European
Society of Cardiology (ESC) and endorsed by the European Society of
Anaesthesiology (ESA). Eur Heart J 30:2769–2812.
Reynolds JC, Callaway CW, El Khoudary SR, Moore CG, Alvarez RJ, Rittenberger JC. (2009).
Coronary angiography predicts improved outcome following cardiac arrest:
propensity-adjusted analysis. J Intensive Care Med 24:179–86.
Spaulding CM, Joly LM, Rosenberg A, et al. (1997). Immediate coronary angiography in
survivors of out-of-hospital cardiac arrest. N Engl J Med 336:1629–33.
Stub D, Hengel C, Chan W, Jackson D, Sanders K, Dart AM, Hilton A, Pellegrino V, Shaw
JA, Duffy SJ, Bernard S, Kaye DM. (2011). Usefulness of cooling and coronary
catheterization to improve survival in out-of-hospital cardiac arrest. Am J Cardiol.
107(4):522-7. Epub 2010 Dec 22.
Sunde K, Pytte M, Jacobsen D, et al. (2007) Implementation of a standardised treatment
protocol for post resuscitation care after out-of-hospital cardiac arrest. Resuscitation
73:29— 39.
Taylor AJ, Cerqueira M, Hodgson JM, et al. (2010).
ACCF/SCCT/ACR/AHA/ASE/ASNC/NASCI/SCAI/SCMR 2010 appropriate
use criteria for cardiac computed tomography: a report of the American College of
Cardiology Foundation Appropriate Use Criteria Task Force, the Society of
Cardiovascular Computed Tomography, the American College of Radiology, the
American Heart Association, the American Society of Echocardiography, the
American Society of Nuclear Cardiology, the North American Society for
Cardiovascular Imaging, the Society for Cardiovascular Angiography and
Interventions, and the Society for Cardiovascular Magnetic Resonance J Am Coll
Cardiol 56:1864-1894.
Thompson PL. (2010). The invasive approach to acute coronary syndrome: true promise or
false premise? Med J Aust; 192 (12): 694-695.
Vahanian A, Baumgartner H, Bax J, Butchart E, Dion R, Filippatos G, Flachskampf F, Hall R,
Iung B, Kasprzak J, Nataf P, Tornos P, Torracca L, Wenink A. (2007). Grupo de
Trabajo sobre el Tratamiento de las Valvulopatías de la Sociedad Europea de
68 Advances in the Diagnosis of Coronary Atherosclerosis

Cardiología: guidelines on the management of valvular heart disease. Eur Heart J.


28: 230–268.
Wijeysundera HC, Vijayaraghavan R, Nallamothu BK, et al. (2007). Rescue angioplasty or
repeat fibrinolysis after failed fibrinolytic therapy for ST-segment myocardial
infarction: a meta-analysis of randomized trials. J Am Coll Cardiol. 49:422–30
Wolfrum S, Pierau C, Radke PW, Schunkert H, Kurowski V. (2008). Mild therapeutic
hypothermia in patients after out-of-hospital cardiac arrest due to acute STsegment
elevation myocardial infarction undergoing immediate percutaneous coronary
intervention. Crit Care Med 36:1780–6
4

History of Coronary Angiography


Ryotaro Wake, Minoru Yoshiyama et al.*
Osaka City University Graduate School of Medicine,
Osaka City General Hospital, Tsukazaki Hospital
Japan

1. Introduction
Diagnostic cardiac catheterization is recommended whenever it is clinically important to
define the presence of cardiac disease that cannot be evaluated adequately by noninvasive
techniques. Because the risk of a major complication from cardiac catheterization is less than
1% with mortality of less than 0.08%, many doctors can perform the catheterization safely
now. We discuss the history of coronary angiography.

2. X-ray
We must talk about X-rays, at first, when we discuss the history of coronary angiography.
In 1895, Roentgen discovered the x-rays. In 1896, X-rays of fractures had been obtained and
published. Walter B. Cannon observed the movements of the opaque mass in the stomach
and subsequently with mixing bismuth subnitrate described the nature and site of peristaltic
activity in cats as seen on the fluoroscopic screen.
Walsh published “The Röntgen Rays in Medical Work. New York: William Wood, 1898.” He
reproduced a chest film that clearly showed the position and dimensions of the heart and
describe a number of cardiac and aortic conditions that he visualized radiographically. The
radiograph in his book required an exposure time of 20 minutes, with the tube only 30
inches (76 cm) from the radiographic plate (Figure 1).

3. Angiogram
Visualization of human blood vessels was achieved in January 1896, during the month after
the announcement of Roentgen’s discovery, when Haschek and Lindenthal injected
Teichmann’s mixture, composed mainly of calcium carbonate, into the blood vessels of an
amputated hand. A published photograph of their original roentgenogram clearly showed
the potential of the method for visualizing the vascular bed (Figure 2).
Morton who was a pioneer American radiologist wrote a text entitled “The X-ray, or,
Photography of the Invisible and Its Value in Surgery. New York: American Technical

* Hidetaka Iida, Hiroaki Takeshita, Takanori Kusuyama, Hitoshi Kanamitsu, Hideya Mitsui,

Yukio Yamada, Shinichi Shimodozono and Kazuo Haze


Osaka City University Graduate School of Medicine, Osaka City General Hospital, Tsukazaki Hospital, Japan
70 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 1. The chest film from “The Röntgen Rays in Medical Work”.
Book Company. 1896.” In this book, his remarks concerning contrast studies are
intriguing.
In teaching the anatomy of the blood vessels, the X-ray opens out a new and feasible
method. The arteries and veins of dead bodies may be injected with a substance opaque to
the X-ray and thus, their distribution may be more accurately followed than by any possible
dissection. The feasibility of this method applies equally well to the study of the structures
and organs of the dead body. To a certain extent, therefore, X-ray photography may replace
both dissection and vivisection, and in the living body, the location and size of a hollow
organ may be ascertained by causing the subject to drink a harmless fluid more or less
opaque to the X-ray, or an effervescing mixture which will cause distention, and then taking
the picture.
In 1920, a radiographic atlas devoted only to the systemic arteries of the body was published
in England, “The X-ray atlas of the systemic arteries of the body. London: Balliere, Tindall,
and Cox, 1920,” by Orrin. The reproductions showed blood vessels in cadavers with great
clarity (Figure 3).
In 1910, Franek and Alwens introduced a suspension of bismuth and oil into the hearts of
dogs and rabbits directly through the large veins and observed the passage of droplets from
the heart into the lungs. In 1922, the work of Sicard and Forestier represented the next major
History of Coronary Angiography 71

advance, when they used Lipiodol, an early oil-based contrast medium, to study the
bronchial tree and then the spinal subarachnoid space. In 1923, they injected 5 ml of
Lipiodol into the femoral vein of a dog and, with the aid of fluoroscopy, watched droplets
move with increasing speed from the iliac vein into the heart. The Lipiodol was then
pulverized by ventricular contraction, thrown with great speed into the pulmonary artery
and finally spread as multiple emboli into the small vessels of the lungs, disappearing in 10-
12 minutes. Nextly, they repeated the experiment with human subjects, in whom they
carefully observed the course of the opaque oil from the antecubital vein to the pulmonary
capillaries. They reported that the patients coughed as the oil reached the lungs but suffered
no other ill effects.
In 1923, Berberich and Hirsch reported the first arteriograms and venograms obtained in
human subjects, using 20% strontium bromide (Figure 4). In 1924, Brooks described the
intraarterial injection of sodium iodide as a means of showing vessels of the lower
extremities in humans (Figure 5).
In 1928, Moniz et al. described carotid angiography and its application to the study of
cerebral lesions (Figure 6).

Fig. 2. A roentgenogram made by Haschek and Lindenthal after the injection of Teichman’s
mixture into the blood vessels of an amputated hand (Haschek E. Lindenthal O’F. A
contribution to the practical use of the photography according to Röntgen. Wien Klin
Wochenschr 1896:9:63.).
72 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 3. Reproduction of a 1920 roentgenogram of a cadaver after the injection of opaque


medium into the cerebral vessels (Orrin HC. The x-ray atlas of the systemic arteries of the
body. London: Balliere, Tindall, and Cox,1920.).
History of Coronary Angiography 73

Fig. 4. Venographic studies obtained by Berberich and Hirsch in 1923. They injected 20%
strontium bromide into the veins of the upper extremity (Berberich J Munchen Klin
Wochenschr 1923;49:2226).
74 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 5. The first femoral arteriogram obtained in a human subject. Brooks used sodium
iodide as a means of demonstrating the vessels of the lower extremities in 1924(Brooks B.
Intraarterial injection of sodium iodide. JAMA 1924; 82:1016.).
History of Coronary Angiography 75

Fig. 6. Reproduction of a cerebral arteriogram (Moniz E. J Radiol Electrol Med Nucl


1928;12:72.)

4. Cardiac catheterization
As Andre Cournand remarked in his Nobel lecture of Decenber, 11th, 1956
(http://nobelprize.org/nobel_prizes/medicine/laureates/). Cournand and his colleagues
led us into a new era in the understanding of cardiac function in humans. According to
Cournand, cardiac catheterization was performed and so named by Claude Bernard in
1844. The subject was a horse, and both the right and left ventricles were entered by
retrograde approach from the jugular vein and carotid artery.
His careful application of scientific method to the study of cardiac physiology using the
cardiac catheter demonstrated the enormous value of this technical innovation. An era of
investigation of cardiovascular physiology in animals then followed, resulting in the
development of many important techniques and principles (pressure manometry, the Fick
cardiac output method), which awaited direct application to the patient with heart
disease.
76 Advances in the Diagnosis of Coronary Atherosclerosis

In 1928, Werner Forssmann, having practiced on a cadaver, inserted a 65 cm catheter into his
own antecubital vein until he felt that it had reached the right atrium [1] (Figure 7). With the
catheter dangling from his arm, he walked through a hospital basement to the radiographic
room. There he obtained a roentgenogram that confirmed his belief that the catheter tip had
in fact reached the right atrium.
During the next 2 years, Forssmann continued to perform catheterization studies including
six additional attempts to catheterize himself. Bitter criticism, based on an unsubstantiated
belief in the danger of his experiments, caused Forssmann to turn his attention to other
concerns, and he eventually pursed another catheter-related career as a urologist.
Nevertheless, for his contribution and foresight he shared the Nobel Prize in Medicine with
Andre Cournand and Dickinson Richards in 1956. Forssmann’s primary goal in his
catheterization studies was to develop a therapeutic technique for the direct delivery of
drugs into the heart.
He wrote, “If cardiac action cases suddenly, as is seen in acute shock or in the heart disease,
or during anesthesia or positioning, one is forced to deliver drugs locally. In such cases the
intracardiac injection of drugs may be life saving. However, this may be a dangerous
procedure because of many incidents of laceration of coronary arteries and their branches
leading to cardiac tamponade, and death. Because of such incidents, one often waits until
the very last moment and valuable time is wasted. Therefore I started to look for a new way
to approach the heart, and I catheterized the right side of the heart through the venous
system.”
Others, however, appreciated the potential of using Forssmann’s technique as a diagnostic
tool. In 1930, Klein reported 11 right heart catheterizations, including passage to the right
ventricle and measurement of cardiac output using Fick’s principle. In 1932, Padillo and
coworkers reported that right heart catheterization and measurement of cardiac output in
two subjects. Except for these few early studies, application of cardiac catheterization to
study the circulation in normal and disease states was fragmentary until the work of Andre
Cournand and Dickinson Richards, who separately and in collaboration produced a
remarkable series of investigations of right heart physiology in humans [2]. In 1947, Dexter
reported his studies on congenital heart disease and passed the catheter to the distal
pulmonary artery, describing “the oxygen saturation and source of pulmonary capillary
blood” obtained from the pulmonary artery wedge position [3]. Subsequent studies from
Dexter’s laboratory [4] and by Werko elaborated the use of this pulmonary artery wedge
position and reported that pressure measured at this position was a good estimate of
pulmonary venous and left atrial pressure. During this exciting early period, catheterization
was used to investigate problems in cardiovascular physiology by McMichael and Sharpey-
Shafer in England. Lenegre and Maurice in Paris and Warren, Stead, Bing, Dexter,
Cournand and others in the United States [5-9].
Further developments came rapidly in the 1950s and 1960s. Retrograde left heart
catheterization was first reported by Zimmerman and others [10] and Limon-Lason and
Bouchard in 1950. The percutaneous (rather than cut-down) technique was developed by
Seldinger in 1953 and was soon applied to cardiac catheterization of both the left and right
heart chambers [11]. Trans-septal catheterization was first developed by Ross [12] and
Cope [13] in 1959 and quickly became accepted as a standard technique. Selective
coronary angiography was reported by Sones and others in 1959 and was perfected to
History of Coronary Angiography 77

remarkable excellence over the ensuing years [14]. Coronary angiography was modified
for a percutaneous approach by Ricketts and Abrams [15] in 1962 and Judkins [16] in 1967.
In 1970, Swan and Ganz introduced a practical balloon-tipped, flow-guided catheter
technique enabling the application of catheterization outside the laboratory [17]. Better
radiographic imaging techniques and less toxic radiographic contrast agents have been
developed progressively, as many numbers of diagnostic catheterizations has performed
(Figure 8).
In 1977, Grüntzig and others introduced the technique of balloon angioplasty, known as
percutaneous transluminal coronary angioplasty (PTCA) [18].

Fig. 7. The first documented cardiac catheterization. At age 25, while receiving clinical
instruction in surgery at Eberswalde, Werner Forssmann passed a catheter 65 cm through
one of his left antecubital veins until its tip entered the right atrium. He then walked to the
radiology department where this roentgenogram was taken (Klin Wochenschr 1929;8:2085.
Springer-Verlag, Berlin, Heidelberg, New York).
78 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 8. Normal coronary angiogram. Left panel: left coronary artery. Right Panel: right
coronary artery.

5. Equipments for coronary angiography


In the l960s, great strides were made in the quality of image amplification, particularly with
the introduction of the cesium iodide tube. Simultaneously, powerful generators capable of
millisecond exposures and high contrast levels were developed. Cine cameras evolved into
dependable instruments with fine-quality lenses that provide extraordinarily good
resolution in modern 35-mm cine angiocardiography. Videotape recording techniques
improved strikingly. Instantaneous playback of tape-recorded images has afforded great
utility and safety in determining whether desired anatomic and physiologic information has
been attained, thereby precluding additional unnecessary studies (Figure 8).
The introduction of angled views for the study of congenital and acquired heart disease was
an important technical innovation [19]. Equipment companies responded with engineering
advances that made angled views relatively simple: the C-arm, the U-arm, and the
parallelogram.
Recently, the development of non-invasive angiography device is advancing.
Echocardiography, scintigraphy, cardiac computed tomography and cardiac magnetic
resonance imaging play increasingly important roles in the evaluation of the heart and great
vessels, non-invasively. It is already an established technique for the evaluation of
congenital heart disease, cardiac tumors and disorders of the aorta and pericardium. They
can evaluate ventricular function and regurgitation of blood across heart valves.

6. Conclusion
Medicine moves so fast that its evolution tends to be forgotten. In particularly, techniques
that have become an integral part of the standard device of diagnosis or therapy are passed
History of Coronary Angiography 79

to each new generation of physicians and trainees. We perform them without their having
sense of efforts involved in the initial development of those techniques.
In this one decade, we would be astonished at how far the field has come and how much
information can be gathered on the structure and function of the heart from diagnostic
imaging. A century is a short time in the history of medicine, but it has been an eventful
lifetime.

7. References
[1] Shapiro, S.M. et al. (1997) Primary pulmonary hypertension: improved long-term effects
and survival with continuous intravenous epoprostenol infusion. J Am Coll Cardiol,
Vol.30, No.2, pp. 343-349
[2] Cournand, A. et al. (1945) Measurement Of Cardiac Output In Man Using The Technique
Of Catheterization Of The Right Auricle Or Ventricle. J Clin Invest, Vol. 24, No.1,
pp. 106-116
[3] Dexter, L. et al. (1947) Studies Of Congenital Heart Disease. Ii. The Pressure And Oxygen
Content Of Blood In The Right Auricle, Right Ventricle, And Pulmonary Artery In
Control Patients, With Observations On The Oxygen Saturation And Source Of
Pulmonary "Capillary" Blood. J Clin Invest, Vol. 26, No. 3, pp. 554-560
[4] Hellems, H.K. et al. (1949) Pulmonary capillary pressure in man. J Appl Physiol, Vol. 2,
No. 1, pp. 24-29
[5] Bing, R.J. et al. (1947) Catheterization of the coronary sinus and the middle cardiac vein
in man. Proc Soc Exp Biol Med, Vol. 66, No. 1, pp. 239
[6] Bing, R.J. et al. (1947) Physiological studies in congenital heart disease. Bull Johns Hopkins
Hosp, Vol. 80, No.2, pp. 107-120
[7] Burchell, H.B. et al. (1948) Cardiac catheterization in the diagnosis of various cardiac
malformations and diseases. Mayo Clin Proc, Vol. 23, No. 22, pp. 481-487
[8] Stead, E.A., Jr. and Warren, J.V. (1947) Cardiac output in man; an analysis of the
mechanisms varying the cardiac output based on recent clinical studies. Arch Med
Interna, Vol. 80, No. 2, pp. 237-248
[9] Stead, E.A., Jr. et al. (1948) Cardiac output in congestive heart failure; an analysis of the
reasons for lack of close correlation between the symptoms of heart failure and the
resting cardiac output. Am Heart J, Vol. 35, No.4, pp. 529-541
[10] Zimmerman, H.A. et al. (1950) Catheterization of the left side of the heart in man.
Circulation, Vol. 1, No. 3, pp. 357-359
[11] Seldinger, S.I. (1953) Catheter replacement of the needle in percutaneous
arteriography; a new technique. Acta radiol 39 (5), 368-376
[12] Ross, J., Jr. (1959) Transeptal left heart catheterization: a new method of left atrial
puncture. Ann Surg, Vol. 149, No. 3, pp. 395-401
[13] Cope, C. (1959) Technique for transseptal catheterization of the left atrium; preliminary
report. J Thorac Surg, Vol. 37, No. 4, pp. 482-486
[14] Ryan, T.J. (2002) The coronary angiogram and its seminal contributions to
cardiovascular medicine over five decades. Circulation, Vol. 106, No. 6, pp. 752-756
[15] Ricketts, H.J. and Abrams, H.L. (1962) Percutaneous selective coronary cine
arteriography. JAMA, Vol. 181, pp. 620-624
[16] Judkins, M.P. (1967) Selective coronary arteriography. I. A percutaneous transfemoral
technic. Radiology, Vol. 89, No.5, pp. 815-824
80 Advances in the Diagnosis of Coronary Atherosclerosis

[17] Swan, H.J. et al. (1970) Catheterization of the heart in man with use of a flow-directed
balloon-tipped catheter. N Engl J Med, Vol. 283, No. 9, pp. 447-451
[18] Gruntzig, A.R. et al. (1979) Nonoperative dilatation of coronary-artery stenosis:
percutaneous transluminal coronary angioplasty. N Engl J Med, Vol. 301, No. 2, pp.
61-68
[19] Bargeron, L.M., Jr. et al. (1977) Axial cineangiography in congenital heart disease.
Section I. Concept, technical and anatomic considerations. Circulation, Vol. 56, No.
6, pp. 1075-1083
5

Coronary Angiography -
Physical and Technical Aspects
Maria Anna Staniszewska
Medical University of Lodz, Department of
Medical Imaging Techniques
Poland

1. Introduction
Diseases of coronary system and heart are the main reason of deaths in highly civilized
population. Then the appropriate diagnostic methods and treating of early disorders are
very important for a population health care. As the imaging techniques play a great role in
good diagnosis they are intensively used and developed. Among the most commonly used
methods is coronary angiography, which involves coronary vessels visualization during
exposure to x-rays, after filling them up with contrast agents.
There are two versions of the procedure:
CCA – conventional coronary angiography, performed under control of x-ray unit with C-arm,
CTA - CT angiography, performed under control of computerized tomography (CT scanner).

2. CCA
2.1 Description of the procedure
A patient lies in a supine position on the table which is a part of the C-arm unit. The x-ray
tube moves rotationally in two perpendicular planes (horizontal and vertical); it makes
possible any needed type of projection. (The most common projections are: LAO,
LAO/Cranial, LAO/Caudal, RAO). The principal current-voltage parameters are chosen
automatically.
Good practice rules require keeping the x-ray tube under the table. This allows avoiding an
unnecessary irradiation of the staff and makes the doses to a patient lower.
To visualize coronary vessels a contrast agent is administered intravenously. intravenously:
first a thin catheter is introduced the brachial artery or the femoral artery (there are two
alternative access routes) and its movement is traced by fluoroscopy and observed on
monitor by the operator.
(Physiological parameters of a patient of patient are permanently monitored during the
procedure.)
The operating team consists of 3 to 4 persons: usually there are an operator (with assistant if
necessary), instrumenting nurse and anaesthetic nurse. All the staff remind around the
patient table and thus may be exposed to quite high doses of radiation. (There are mainly
scattered x-rays). The most exposed member of the team is the operator: the doses registered
for such persons may achieve the highest level measured for occupational exposures [1,2].
82 Advances in the Diagnosis of Coronary Atherosclerosis

Patient exposure results mainly from the primary x-ray beam which covers a part of the
back surface (the area of left shoulder) by the most time of the procedure. The remaining
parts of the patient’s body are exposed rather to scattered radiation.
The images created during the procedure can be registered in three ways:
a. Real-time images (fluoroscopic images) observed only by the operator,
b. Some chosen moments of the procedure recorded in radiography mode (so-called
acquisition),
c. Long periods of exposure can be recorded in so-called “cine-mode” (if this mode is
implemented in the x-ray system).
The results of CCA procedure are analyzed by cardiologists during its performance and also
retrospectively afterwards.
Most of x-ray units used nowadays in cardiology are equipped with the digital image
recording system. They are two types of them: (1) image intensifier based systems and (2)
flat-panel fluoroscopy systems.
The first one uses conventional technology: the output screen of image intensifier is
projected in a video camera or a CCD camera to produce an electronic information. (In the
previously used conventional image intensifier analogue system the output screen had been
projected on the film by optical lens).
In the flat-panel systems signals created by x-rays are electronic and are thus recorded
simultaneously.
(The fundamental difference between “analogue” and “digital” systems concerns to way of
image archiving (i.e. film or electronic information). The both systems produce the “real-
time” image, although the flat-panel systems create the image by dividing the primarily
detected signal. That is why the modern x-ray system has much higher output capacity and
thus higher doses are possible).

2.2 Technical requirements for x-ray systems used for CCA


X-ray system used for CCA procedures should be equipped as follows:
- constant potential generator of minimum 80 kW (in possibly rectangular pulses),
- x-ray tube focuses 1.2/0.5mm,
- overcouch image detector,
- distance between x-ray tube and image detector is tracked (minimum focus skin
distance 30 cm),
- concave couch top for patient comfort (made from low-attenuating materials),
- dose-area product meter (display visible for operator),
- staff protective shielding.
The system should also have the following additional tools and fulfil the following
requirements:
- display of fluoroscopy time, total dose-area product (fluoroscopy and radiographic)
and estimated skin entrance dose,
- minimum two dose-rate options (Low-Medium-High or Low- Standard),
- additional filtration (Cu preferred) and collimators incorporating circular shutters,
- flexibility of pulsed fluoroscopy mode,
- image hold system,
- possibility of AEC mode choice by the user by the user (IMAGE or DOSE weighted).
Additionally in some x-ray systems there are also available partially absorbent contoured
filters to control the bright spots produced by the lung tissue bordering the heart.
Coronary Angiography - Physical and Technical Aspects 83

2.3 X-ray dosimetry in CCA


The quantities used to assess radiation risk in interventional radiology are following:
Entrance air kerma (EAK), surface dose , dose-area product (DAP), absorbed organ dose,
effective dose.
EAK, surface dose and DAP are available for measurements. The remaining can be evaluated
experimentally or theoretically.
EAK and surface dose can be measured directly (using appropriately calibrated devices) or
indirectly (by thermoluminescence (TL) dosimeters or film badges). The indirect methods
need a calibration of dosimeters by the comparison to a reference meter for the standardized
exposure (in a competitive laboratory). These methods are routinely applied to individual
dosimetry of occupationally exposed persons, but are also used for patients in radiological
procedures. (TL dosimeters are preferred for the staff in interventional procedures.)
X-ray systems used for CCA are obligatory equipped with a DAP-meter to summarize the
emission in all modes of work (i.e. fluoroscopy, radiography, cine). (Dose Area Product=
DAP [mGy.cm2]). Measurements are performed by the transmission chamber placed on the
x-ray tube output.
Absorbed organ doses are necessary for evaluation of a possible effect for an exposed human.
They can be estimated theoretically or experimentally. Theoretical evaluation relies on
Monte Carlo simulation performed for mathematical human phantom at assumed exposure
conditions. In the experimental method the physical anthropomorphic phantom is used
which is exposed at assumed exposure conditions while the dosimeters are placed inside.
(TL dosimeters are used usually for this purpose.)
On the basis of the absorbed organ doses the evaluation of the equivalent dose and the
effective dose are possible [3,4].
Equivalent dose (in an organ or body part)
Ti=wR.Di [Sv] where wR-the radiation weighting factor (wR=1 for x-rays, γ-rays and
electrons), Di- the dose absorbed in “i” organ (or body part).
Effective dose (whole body)
Eff= ∑wi.Di [Sv] had been implemented to evaluate overall risk for health resulted from
exposure to ionizing radiation. This value is fully justified if irradiation of the body is
uniform enough: if opposite deterministic effects can appear despite the very low value of
the Eff .
Both quantities (Ti and Eff) are used for the dose limits applied for occupational exposure.

2.4 Doses to patients


The doses obtained by the patients undergoing CCA procedures are dependent on:
- patient architecture (BMI-body mass index),
- emission effectiveness of the x-ray system,
- applied dose-rate mode,
- total exposure time,
- mode of work (number of acquisitions -radiographic or cine).
The entrance surface dose to patient dramatically increased when the focus-to-skin distance
becomes too short.
The patient doses are significantly higher when the high dose-rate mode is activated or if
pulsed fluoroscopy of high number of pulses per second is chosen. These doses are also
84 Advances in the Diagnosis of Coronary Atherosclerosis

inversely proportional to the diameter of the image intensifier. The entrance dose to skin
may locally achieved values exceeding the threshold of deterministic effects and the skin
injuries of patients undergoing interventional procedures are reported [5].
Generally, exposure of the patients during CCA procedures is extremely non-uniform:high
doses are received on a small are of the back while the remaining parts of the body
practically stay out of the primary beam. Although effective dose is not a good measure of
radiation risk for that, this quantity is used after all. The measurable quantities, i.e. entrance
surface dose (or air kerma) and DAP, have more practical meaning. These quantities are
proportional to each other:

DAP = EAKc. S
where EAKc –the entrance air kerma in the centre of the beam, S – mean value of incident x-
ray beam are (on patient’s skin).
(Both values are averaged over the total exposure time, because their varies from one
projection to another.)
DAP (and EAKc) values have to be displayed on the monitors of x-ray system and should be
registered in the patient record. (Especially when EAKc>1Gy and exceeds the threshold of
deterministic skin effects.)
As the doses to patients in CCA are affected by a number of factors the range of their values
is very wide. The examples are presented in Table 1 and Table 2.
To keep the radiation risk to patients on an acceptable level and control it the reference
levels are introduced also for interventional cardiology procedures (like for another
radiological procedures).

Number of Time of Number of


DAP [Gy.cm2] Reference
patients fluoroscopy [min] frames
130 72 +/-55 0.35+/-0.25 1550+/-775 [7]
78 73 9.9 1079 [8]
100 60.6 - 412 [9]
117 1.1 – 11.3 0.3 - 22 - [10]
8 22 - 93 2 - 21 Doctor 1 [11]
3 14 - 44 1–4 Doctor 2
106 35 - 160 4.8+/-3.5 39+/-11 [12]
62 37-190 4.2+/-3.0 30+/-10
90 3.1-57.2 1.5-5.1 639 [13]
76 44.5 6 10 [14]
194 64 - 281 0.4 - 33 200-1911 [6]
Table 1. The doses to patients in CCA procedures (according to [6])
Effect of pulsed fluoroscopy on the patient dose in CCA were investigated by [16]:
X-ray systems with pulsed fluoroscopy (5 vendors): 10 units; average surface dose rate:
22.52+/-4.5 mGy/min, (25, 30 and 50 pulses/sec);
X-ray systems with conventional fluoroscopy (3 vendors): 3 units; average surface dose rate:
23.93+/-2.77 mGy/min.
Coronary Angiography - Physical and Technical Aspects 85

DAP [Gy.cm2] Time of fluoroscopy [min]


X-ray system
median range median range
CCD 30.0 1.1-135.2 3.8 1.1 – 57.0
FP 31.0 14.7-75.9 4.0 0.8- 39.4
Table 2. Median and range of DAP and fluoroscopy time in CCA for different x-ray
systems [15]

2.5 Doses to staff


The main inconvenience of CCA procedures (generally – interventional radiology) is the
necessity to work standing near the patient during exposure, i.e. in the radiation field. Thus
the staff is also exposed mainly to scattered radiation, but primary x-rays are also possible.
The factors affecting staff dose are as follows:
- relative position with respect to patient,
- patient body typ (BMI),
- irradiated patient volume,
- x-ray tube position,
- time of exposure,
- effective use of protection shielding.
Thus, staff and patient doses are partially linked: higher exposure for patient means higher
irradiation to staff. This is especially true for an operator and the person standing nearby
him (an assistant or instrumenting nurse). Additional factor determining the doses to staff is
an professional experience and good training in the procedure performance: good manual
skills make the exposure time shorter.
Patients with high BMI need a high dose-rate to produce the diagnostic image and then the
scattered x-rays also achieve high intensity (especially near the patient).
X-ray tube in undercouch position allows to reduce the doses (mainly for eyes and thyroid
of the operator) and also allows avoiding the contact with primary x-rays. (The old x-ray
systems had the tube in overcouch position which caused very high doses to operator’s
hands).
The problem of radiation risk to staff in interventional cardiology had been widely
discussed many times. The resulting essential conclusions are the recommendations
concerning performance of the interventional procedures and elaboration of the
methodology for evaluation radiation risk for the staff [1,17].
The main recommendations concerning the proceeding of the staff during CCA can be
summarized in the three points:
- time of exposure possibly short,
- distance to patient possibly long,
- protection shield used effectively.
The recommendations concerning radiation risk rely upon evaluation of the doses to the
staff.
Because of the spatial distribution of x-rays affecting staff exposure the special dosimetric
requirements are introduced for this group of workers: they should have been equipped
with minimum two dosimeters, one under the protective apron (at the chest level) and the
second over the apron at the collar level. The first dosimeter is used to evaluate the effective
dose and the second can be used to evaluate the equivalent dose to the eyes. (Also for the
thyroid if it is not shielded.) Additional dosemeter(s) can be use to evaluate the equivalent
86 Advances in the Diagnosis of Coronary Atherosclerosis

dose(s) to the hands. The protective aprons used by the staff in interventional procedures
should have Pb-equivalent of 0.35mm.
As the doses to staff in CCA are affected by a number of factors the range of their values is
very wide (the same concerns patients) and many studies were performed to evaluate them.
The range of the doses shows the results given in the paper from the USA [18], where on
the basis of the rich collection of published data the exposure for operators in
cardiovascular procedures from the early 1970’s through the present was analyzed. The
range of effective doses from 0.02μSv to 38 μSv per one cardiac diagnostic catheterization
procedure was found.
Another survey had been performed by European research group SENTINEL in a sample of
European cardiac centres and the results have been published recently [19]. According to
this study the results for the first operator are as follows:
a. annual effective dose: median 1.3mSv, third quartile 1.4mSv,
b. equivalent dose over the apron: median 11.1mSv, third quartile 14mSv.
Although the above values are rather low it should be underlined that they may be many
times higher (even up the annual limits) , especially for inexperienced staff or in a case of
clinically difficult patient.

3. CTA
A very dynamic development computerized tomography (CT) Which occurred during last
decade creates quite new possibilities for this imaging method. The rotation time below one
second together with a rich software allow applying CT to diagnostics of dynamic
physiological processes and quickly moving objects (organs). On the other hand, increasing
frequency of heart diseases in population linked the health needs With technological
achievements in the CT technique to implement this imaging method to cardiology.
Although traditional (conventional) coronary x-ray diagnostics remain a gold standard
these procedures are invasive and resulted in high radiation doses both to patients and to
staff. Therefore CT procedures became an alternative and comfortable method for heart
diagnosis.
For technological reasons the possibilities of CT are limited to diagnostics, without further
therapeutic activity (i.e. angioplasty). However, cardiovascular CT plays an increasingly
important role in planning for cardiovascular interventions (those are performed under C-
arm x-ray units).
Improvements in spatial and temporal resolution, scan time, scan range and advanced
image postprocessing (very important in clinical practice) have made CT angiography (CTA)
an excellent tool for identifying patients in need of invasive therapy and for mapping out
the best percutaneous or surgical approach. In some cases CTA provides complementary
information to conventional angiography (CCA).
As coronary disease is jointed with calcification of coronary arteries evaluation of this
process is a good indicator of pathology. CTA gives possibility to display calcifications (3D)
and to assess them quantitatively: it is so-called “calcium scoring”.
Although history of CTA had begun with four-slice scanners, the real development came
with 64-slice scanners with appropriate combination of spatial and temporal resolution.
Generally, the heart can be imaged well if its structures do not move significantly during the
scanning time. This condition is fulfilled during the diastolic phase of cardiac cycle. For
Coronary Angiography - Physical and Technical Aspects 87

selection of the proper phase for CT images recording ECG gating (triggering) is applied.
Additionally, to collect sufficient image data the table speed should not exceed v= SC . fH
and pitch should not exceed p=fH. trot,
where SC- total collimation width (CT primary beam), fH- frequency of heart, trot- rot1ation time.
(For example: if trot=0.5sec and fH=60bpm then p≤0.5 )
The main benefit from CTA is the evaluation of cardiac structure and function. That
concerns coronary anomalies, right and left ventricular function, left ventricular ejection
fraction and myocardial viability assessment, when the results of other modalities are
inadequate [1,2].
According the American College of Cardiology Foundation (ACCF) the principal
indications to cardiac CT are as follows [3]:
- low and intermediate pretest probability of obstructive coronary artery disease (CAD),
- noncontrast CT calcium scoring for patients at intermediate risk of coronary heart
disease, and for low-risk patients with a family history of disease,
- coronary CT is especially recommended for patients with reduced left ventricular
ejection fraction at low or intermediate pretest probability of disease,
- preoperative CT angiography for both heart surgery and noncoronary indications in the
setting of risk of CAD.

3.1 Description of the procedure


General rules for exposure parameters:
Slice thickness <1mm, pitch<1, trot≤0.5sec, kilovoltage U≥100kV, mAs and physical filtration
are dependent on the CT scanner, ECG-gating.
The main stages of CT study protocol are as follows:
- The heart rate control and regulation (if necessary: should have been lower than
65bpm),
- Calcium scoring,
- i.v. injection of contrast agents,
- CT angiography.
Ad.1. The required heart rate is dependent on the scanner software: at the high cardiac rate
the registered imaging data also have to be reconstructed very quickly (practically
immediately). This is possible only with the newest scanners. Lowering the heart rate is
achieved pharmacologically.
Ad.2. Calcium scoring is performed on the basis of pre-contrast images. The high
attenuation of x-ray in calcified structure allows to differentiate them from the soft tissues.
Ad.3. Contrast agent (iodine compound) is injected automatically in the rate adapted to data
acquisition.
Ad.4. CT coronary angiogram: the data acquisition starts automatically when the signal in
the descending aorta reaches a predefined threshold (usually 100 HU). The entire volume
of the heart is scanned during a few seconds: the newest versions of CT scanners (≥64-
slice) do it during one breath-hold (<10sec) under ECG control. The tube voltage, anode
current and gantry rotation time are dependent on the scanner type and model and
version of software.
After completing acquisition of primary data, elaboration of them (post-processing) begins.
The software dedicated to coronary procedures is necessary for that. Such software is
offered by the vendors on user’s requirement.
88 Advances in the Diagnosis of Coronary Atherosclerosis

It should be underlined that imaging possibilities of each CT scanner are determined by its
technical configuration (number of detectors, generator, etc.) but even more by the installed
version of software.
The principal algorithms for image post-processing are [4]:
- curved and multiplanar reformats (MPR),
- shaded-surface display (SSD),
- maximum intensity projection (MIP),
- volume rendering (VR).
The axial images produced by hardware (so-called row data) are essential and should be
used but reformatted images may improve lesion detection and classification (particularly
coronal and oblique views). The axial images and MPRs are used for diagnosis and the SSD
and MIPs are for display purposes.
The results of CTA are evaluated or consulted by a cardiologist.

3.2 Dosimetry in CT
Because of special exposure system CT needs the special dosimetry quantities, i.e.: dose-
length-product (DLP) and computed-tomography-dose- index (CTDI).
CTDI is treated as an estimation of mean absorbed dose to the whole scanned volume. This
quantity also evaluates the level of radiation risk for given procedure at assumed exposure
parameters.
CTDI is calculated automatically by scanner software on the basis the selected exposure
parameters and is displayed on CT monitor. The DLP is then computed as a product of
CTDI (averaged over the whole scan length) and the scan length: such value is displayed on
CT monitor after the examination is completed.
Independently, CTDI can be computed on the basis of measurements performed for particular
dosimetric phantoms. For that purpose DLP is measured using special pencil-shaped
chamber (external probe) to summarize the dose from incident photons during the tube
rotation on the whole length of scanned object. (This is also performed during the
acceptance or constancy Quality Control (QC) tests for CT scanner.)
Final evaluation of risk to patient in CT procedures is based on the effective dose. As in the
other radiological techniques this quantity can be estimated theoretically (Monte Carlo
simulation) or experimentally (TL dosimeters inside the anthropomorphic phantom).
Additionally – the effective dose obtained in the particular procedure can be estimated
indirectly:

Eff= k . DLP
where DLP – value for the procedure, k – transition coefficient for the examined body part
(computed theoretically). The above evaluation is applied for adult patients only.
For coronary examinations the transition coefficient k=0.014 mSv.mGy-1.cm-1 is used which
corresponds to the chest as the examined region [5].
The problem of doses in CTA is considered only for patients as the staff is out of radiation area.

3.3 Doses to patients and radiation protection in CTA


Because of exposure conditions in CTA (thin slices and low pitch) the doses to patients are
relatively high, especially at 64-slice scanners. It was an inspiration to implement the
methods of dose reduction without loss of image quality.
Coronary Angiography - Physical and Technical Aspects 89

The doses can be reduced by lowering of current-voltage parameters, implementation of x-


rays intensity modulation (for three axis) and shortening of scanned body length.
The special solution for dose reduction being dedicated to cardiac CT examinations is
change of image acquisition technique from RGH to PGA [6].
RGH = Retrospectively Gated Helical: the patient and table move through the gantry at a
steady speed. A low pitch (0.2 to 0.4) is needed to cover the entire cardiac volume, especially
to compensate for any ectopic beats, which can result in misregistration and gaps in
coverage. Thus, the same anatomy can be exposed to the x-ray beam many time (up to five)
to ensure enough coverage, causing the high absorbed doses.
PGA= Prospectively Gated Axial: the table is stationary during PGA image acquisition, then
moves to the next location for another scan that is initiated by the next normal cardiac cycle.
The x-rays emission is then dynamically predicted on the basis of ECG signal.
Diagnostic values of images obtained for PGA protocol were not found to be lower for RGH
while the doses where substantially lower [6]. In the same study including 203 CTA exams
(82 with routine RGH and 121 with the PGA) the doses were evaluated for protocol
including scout images, low-dose calcium scoring scans, test-bolus scan and CT coronary
angiogram. The exams were performed by using 64-slice CT scanner (LightSpeed VCT XT,
GE). The effective doses were as follows:
RGH: (8.7-23.2) mSv, mean: 18.4 mSv;
PGA: (0.75-6.67) mSv; mean: 2.84 mSv.
For comparison, the results of the big study involved nearly 2000 patients examined (CTA)
in 50 hospitals in 2007 in Germany and US show the range of effective dose (8-18) mSv with
median 12 mSv. The reduced 100 kV tube voltage was used in 5% of patients, with
sequential scanning in 6%. ECG gating was applied in 73% exams [7].
The influence of lower tube voltage is illustrated also by the results CTA performed using
320-MDCT for anthropomorphic phantom (ECG gating was simulated) [8].
The results are given in Table 3.

Heart rate= 60bpm 100kV 120kV 135kV


Midbreast 13.4±3.6 21.7±4.1 29.3±5.2
Breast 11.8±0.6 18.9±1.1 24.8±1.5
Lung 12.2±2.6 19.1±3.2 26.5±5.0
Thyroid 0.4±0.1 0.7±0.1 0.9±0.2
Heart rate=75bpm 100kV 120kV 135kV
Midbreast 38.3±2.0 59.7±3.1 77.8±3.7
Breast 26.2±1.7 44±1.1 52.8±4.1
Lung 38.0±1.6 58.9±1.7 78±2.9
Thyroid 0.8±0.2 1.0±0.2 2.2±0.5

Table 3. The effect of tube voltage and heart rate on absorbed doses [mGy] in CTA
(according to [8]).
90 Advances in the Diagnosis of Coronary Atherosclerosis

4. Comparing CCA and CTA


Primary x-ray beam in the CCA is focused on nearly the same area of a patient’s body by
most time of the procedure, and then very high local doses are possible. (Skin injuries and
even of x-ray burn cases were reported in the paper [9]).
Moreover, a possibility to destroying the vessel wall during catheter operations also exists.
Patient entrance doses change with body thickness, required image quality and distance to
x-ray tube focus. Because medical staff have to work in direct neighbourhood of x-ray
source and x-rays scattered on a patient’s body, the doses to the staff are related to the
patient doses.
Thus, the procedures are still treated as invasive and risky for both patients and the staff
despite the benefit from “traditional” angiographies.
Good spatial and temporal resolution of modern CT scanners is the reason of positive
features of CTA in comparison to conventional angiography [10,11,12], i.e.:
- better detection of stenosed lesions,
- possibility to delineate a cross-sectional cut of the artery,
- non-invasive procedure contrary to the conventional angiography or intravascular
ultrasound.
All of the above reasons have made CTA an excellent technique for quick (emergency)
cardiac diagnosis and for qualification patients for angioplasty. Moreover, coronary CTA
includes a complete evaluation of the lungs and chest. Contrary to to conventional cardiac
angiography (CCA) the staff are completely unexposed in CTA.
The only deficiency of CTA in comparison to CCA is its exclusive diagnostic character: any
further intervention as angioplasty is not possible in CTA technique. That needs continuing
of treatment action in more “traditional” way – at C-arm unit control.
For comparison of doses to patients in both imaging techniques techniques the
measurements were performed using multislice CT scanners (MSCT) and C-arm units.
The measurements were performed for
- two sixteen-slice CT scanners (made by two manufacturers),
- one sixty-four-slice CT scanner,
- three C-arm units of different manufacturers.
For each controlled C-arm unit the organ doses in “traditional” coronary angiography were
distributed extremely non-uniformly: from up to 100 mGy in the area of the primary x-ray
beam interaction to about zero in quite near vicinity of that. For that in all the phantom
examinations doses for both lungs and doses for elements of skeleton differed significantly
in dependence on the composition of projections.
That was not observed for CTA procedures, when the magnitude of doses inside the
scanned volume was similar.
The detailed results were published [13]. Below the most important values of are presented
in the Tables 4 to 6.
To summarize of the presented results:
1. In coronary angiography carried out by C-arm units patient organ doses are distributed
non-uniformly inside patient body (from up to 100 mGy in area of primary x-ray beam
to about zero in quite near vicinity). Therefore, a low effective dose does not mean the
low absorbed doses at all.
Coronary Angiography - Physical and Technical Aspects 91

2. Doses to patients in CTA procedures covering the chest are distributed more uniformly
and are dependent on the scanner configuration and protocol of the procedure.

CCA CTA
1st manufacturer: 0.9 – 1.4 16-slice scanner: 5.4
2nd manufacturer: 1.3 – 3.2 64-slice scanner: 23.1
(The both scanners made by 1st
3rd manufacturer: 6.2 – 18.8
manufacturer.)

Table 4. Effective doses to patients in coronary procedures [mSv]

Organ Eight exposures carried Four exposures carried


out by C-arm units out by C-arm units the
designed by three same type (made by
manufacturers. the1st manufacturer)

Lungs 2.95 – 23.68 2.95 – 5.64


Stomach 0.07 – 59.76 0.22 – 0.75
Upper Large Intestine <0.1 – 10.73 0.10 – 0.37
Red Bone Marrow 1.75 – 23.90 1.75 – 3.30
Thyroid 0.13 – 2.03 0.13 – 2.03
Heart 0.52 – 27.15 1.39 – 3.75

Table 5. Range of absorbed doses [mGy] in CCA

Organ Sixteen-slice CT Sixty-four-slice CT


Lungs 25.8 64.5
Stomach 5.7 44.2
Upper Large Intestine 1.5 8.1
Red Bone Marrow 10.9 31.4
Thyroid 6.8 11.7
Heart 29.0 77.7

Table 6. Absorbed doses [mGy] to patients in CTA (both CT scanners made by 1st
manufacturer)
92 Advances in the Diagnosis of Coronary Atherosclerosis

5. References
5.1 References for CCA
[1] International Commission on Radiological Protection, Publication 85, Avoidance of
Radiation Injuries from Medical Interventional Procedures, 2001.
[2] Vano E, Gonzales L, Guibelalde E, et al:Radiation exposure to medical staff in
interventional and cardiac radiology. Br.J.Radio., 71,954-960,1998.
[3] International Commission on Radiological Protection, Publication 60, Recommendations
of the International Commission on Radiological Protection, 1991.
[4] International Commission on Radiological Protection, Publication 103, The 2007
Recommendations of the International Commission on Radiological Protection,
2007.
[5] Vano E, Arranz L, Sastre JM, et al: Dosimetric and radiation protection considerations
based on some cases of patient skin injuries in interventional cardiology.
Br.J.Radiol., 71, 510-516, 1998.
[6] Bor D, Olgar T, Toklu T, et al.: Patient doses and dosimetric evaluations in interventional
cardiology. Physica Medica, 25,31-42, 2009.
[7] Maccia C: An optimization study of cardiac interventional radiology procedures: an
experience in France. ERPET in Madrid, p.113-119, 1997.
[8] Hansson B, Karambatsakidou A: Relagtionships between entrance skin dose, effective
dose and dose area product for patients in diagnostic and interventional cardiac
procedures. Radia.Prot.Dosimetry. 90: 141-144, 2000.
[9] Van de Putte S, Verhaegen F, Taeymans Y, et al.: Correlation of patient skin doses in
cardiac interventional radiology with dose-are product. Br.J.Radiol. 73: 504-513,
2000.
[10] Clark AL, Brennan AG, Robertson LJ, et al.:Factors affecting patient radiation exposure
during routine coronary angiography in a tertiary referral centre. Br.J.Radiol. 73:
184-189, 2000.
[11] McFadden SL, Money RB, Shepherd PH: X-ray dose and associated risks from
radiofrequency catheter ablation procedures. Br.J.Radio. 75: 253-265,
2002.
[12] Delichas MG, Psarrakos K, Molyvda Athanassopoulou E, et al.:Radiation doses to
patients undergoing coronary angiography and percutaneuos transluminal
coronary angioplasty. Radiat.Prot.Dosimetry.103(2):149-154, 2003.
[13] Sandborg M, Fransson SG, PetterssonH: Evaluation of patient and staff absorbed doses
during coronary angiography and intervention by femoral and radial artery access
routes. Eur.Radiol. 14(4):653-658, 2004.
[14] Leung KC, Martin CJ: Effective doses for coronary angiography. Br.J.Radiol. 69:426-
431, 1996.
[15] Tsapaki V, Kottou S, Kollaros N, et al.: Dose performance evaluation of a charge
coupled device and a flat-panel digital fluoroscopy system recently installed in
an interventional cardiology laboratory. Radiat.Prot.Dosimetry. 111(3): 297-
304,2004.
Coronary Angiography - Physical and Technical Aspects 93

[16] Chida K, Fuda K, Saito H, et al.: Patient skin dose in cardiac interventional procedures:
conventional fluoroscopy versus pulsed fluoroscopy. Catheter Cardiovasc
Interv.69(1):115-121, 2007.
[17] International Commission on Radiological Protection, Publication 75, General
Principles of Monitoring for Radiation Protection of Workers, 1997.
Kim KP, Miller DL, Balter S, et al.: Occupational radiation doses to operators performing
cardiac catheterization procedures. Health Phys. 94(3):211-227,2008.
[18] Padovani R: Optimisation of patient and staff exposure in interventional radiology. In:
Radiation Protection in Medical Physics, edited by Y.Lemoigne and A.Caner,
Springer in Cooperation with NATO Public Diplomacy Division, The Netherlands,
2011.

5.2 References to CTA


[1] Korosoglou G, Mueller D, Lehrke S, et al.: Quantitative assessment of stenosis severity
and atherosclerotic plaque composition using 256-slice computed tomography.
Eur.Radiol 20:1841-1850,2010.
[2] Rutten A, Meijs MFL, de Vos A, et al.: Biphasic contrast medium injection in cardiac CT:
moderate versus high concentration contrast material at identical iodine flux and
iodine dose. Eur.Radiol. 20: 1917-1925, 2010.
[3] Taylor A, Cerquiera M in Journal of the American College of Cardiology, October 2010:
according to AuntMinnie.com (November 1,2010): New cardiac CT guidelines
expand use for low-risk patients.
[4] W.A.Kalender: Computed Tomography. Publicis MCD Verlag, Munich 2000.
[5] Bischoff B, Hein Fm Meyer T, et al.: Impact of a reduced tube voltage on CT angiography
and radiation dose: results of the PROTECTION I study.JACC Cardiovasc Imaging
2:940-946, 2009.
[6] AuntMinnie.com February 15, 2007: Radiation dose slashed in 64-slice coronary CTA (by
Eric Barnes)
[7] AuntMinnie.com February 3, 2009: JAMA study finds wide variation in cardiac CTA
dose (by Eric Barnes)
[8] Nikoito D, Khosa P, Lin PJ, et al.:Absorbed radiation dose in radiosensitive organs
during coronary CT angiography using 320-MDCT: effect of maximum tube
voltage and heart rate variations. AJR.195(6):1347-1354, 2010.
[9] Vano E, Gelcolea J., Galvan C et.al. Skin radiation injuries in patients following repeated
coronary angioplasty procedures. The British Journal of Radiology 2001; 74: 1023-
1031.
[10] De Backer G, Ambrosioni E, Borch-Johnson K, et al. European quidelines on
cardiovascular disease prevention in clinical practice. Eur.J.Cardiovasc.Prev.
Rehabil. (2003), 10(4):S1-10.
[11] Nieman K, Cademartiri F, Lemos PA, et al.Reliable non-invasive coronary
angiography with fast submillimeter multislice spiral computed tomography.
Circulation 2002, 106: 2051-2054.
94 Advances in the Diagnosis of Coronary Atherosclerosis

[12] Ropers D, Baum U, Polite K, et.al. Detection of coronary artery stenoses with thin-slice
multi-detector row spiral computed tomography and multiplanar reconstruction.
Circulation 2003. 107:664-666.
[13] Staniszewska MA, Jazwinski J: MSCT and C-arm units: doses to patients in the chest
angiography procedures. Pol.J.Radiol. 73(1):43-49, 2008.
6

Procedural Techniques
of Coronary Angiography
Jasmin Čaluk
BH Heart Center Tuzla
Bosnia-Herzegovina

1. Introduction
Coronary angiography is an invasive diagnostic procedure in which radiocontrast is injected
into the coronary arteries under X-ray guidance in order to display the coronary anatomy
and possible luminal obstruction. Despite the advances in other diagnostic methods, it
remains to be "the golden standard" of coronary disease diagnostics. Although today the
complication rate is far lower than previously, the possibility of complication still exists, and
an invasive cardiologist must be able to complete the procedure flawlessly, and to
competently deal with complications, should they occur. In order to be able to do that,
he/she must master the proper techniques in performing the coronary angiography
procedure, and be comfortable with all the available access-sites.

2. Cath lab
The procedure is performed in a specifically designed room, very much resembling the
operating room, called the catheterization laboratory, or cath lab (Fig.1). Recordings are made
using a special X-ray machine, called cardioangiograph (Fig.1). Today, these machines are
completely digitalized, enabling superb picture quality, with the lowest possible radiation
doses, and some additional features, such as precise measurements and quantifications of the
recorded angiograms, connections to additional devices, such as intravascular ultrasound,
easy and large-capacity data storage etc. It is convenient to have a wide choice of spare
catheters, multi-packs, guidewires, coronary angioplasty ballons, coronary stents, and other
equipment in the cath lab itself, so it can be used without time-delay. The space should be
intuitively organized, and the spare devices, sterile gowns, gloves, drapes etc. must be easily
accesible. Defibrillator, endotracheal tubes, temporary pacemakers and leads, oxygen supply,
and other cardiopulmonary resuscitation equipment must be held in the cath lab at all times.
Just outside of the cath lab is the control-station with monitors for angiogram, patient data,
electrocardiogram (ECG), haemodynamics etc (Fig.2). Modern cardioangiographs allow for
data analysis both from the control station outside the cath lab, as well as from within the cath
lab, using specifically designed control-commands at the table.

3. Coronary angiography: Preparation


Coronary angiography is performed under local anesthesia. The procedure is sterile, and all
potential access sites must be disinfected, shaved, and sterilized. At the beginning of the
96 Advances in the Diagnosis of Coronary Atherosclerosis

procedure, the patient lays down in supine position on the cardioangiograph table, and is
prepared for the procedure in sterile conditions (Fig.3).

Fig. 1. Catheterization laboratory

Fig. 2. Control-station with monitors outside the catheterization laboratory


Procedural Techniques of Coronary Angiography 97

Fig. 3. The patient in supine position on the cardioangiograph table


Coronary angiography is performed with the patient in the fasting state, except in
emergency cases, e.g. acute myocardial infarction, in which the degree of urgency prevails
over the risks of aspiration incidents, which are generally very rare. In many institutions
today, it is common not to administer pre-medication prior to coronary angiography, except
in certain specific conditions. In other institutions, the patients are given 10 mg diazepam
p.o. and 25 mg diphenhydramine p.o. 30-60 min before the procedure. If the patient has a
known allergy to radiocontrast, prednisone can be administered at 60 mg p.o. every 8 hours
for two days prior to the procedure. Other institutions have the policy to administer
hydrocortisone 80-120 mg IV some 30 min before the procedure, along with 50 mg
diphenhydramine p.o. The patients with a history of radiocontrast allergy must be
monitored after coronary angiography for signs of acute allergy: urticas on skin or itchy
skin, swelling of tongue, stuffy nose or signs of respiratory distress.
The table set for coronary angiography (Fig. 4) can differ from one institution to another, but
generally it has the following list of items and equipment:
- sterile drapes to cover the patient and radiation-protection screens,
- sterile swabs for cleaning the arterial access site,
- a bowl of Betadine solution for cleaning the arterial access site,
- lidocaine for local anesthesia, 15-20 ml of 2% solution (in some cases, for more sensitive
patients, more may be needed)
- punction neelde for arterial cannulation,
- an 11-mm or 15-mm blade,
- syringes and needles,
- two containers, one with 500 ml of saline solution with 5,000 U of heparin added,
- arterial sheath with a dilator,
98 Advances in the Diagnosis of Coronary Atherosclerosis

- a J-shaped guidewire,
- diagnostic catheters for coronary angiography; start with the most common JL4 and JR4
with angulated pigtail if left ventriculography and/or aortography is planned,
- a three-outlet manifold which is to be connected to pressure transducer, saline solution
container, and radiocontrast container on side-outlets, to the syringe for radiocontrast
application on the proximal end, and with its distal end to diagnostic/therapeutic
catheters during the procedure.
During the procedure, the patient must be monitored by several systems, and the ECG is
absolutely essential. The patient also must have an ECG recording done just before the
procedure actually starts. For practical reasons, during the angiography itself, there is really
no need to apply the precordial leads, since the basic four electrodes can provide enough
information for ECG monitoring during coronary angiography and percutaneous coronary
interventions (PCI). Four electrodes should be placed on the calf or thigh and on the
shoulders or upper arms (Watson & Hunter, 2011).
The area of the puncture site must be shaved, disinfected, then anesthetized, and only then
punctured. The center of the field for shaving and disinfection is the puncture site. The
disinfection is performed starting at the center, and spiraling outwards, at least twice, using
Betadine. Thereafter, the site is wiped off with a third swab. If the procedure is planned to
be performed via the femoral artery, than both right and left groins must be shaved and
disinfected, in case that the operator must switch to contralateral artery. Only after the
disinfection of the puncture site, the patient is covered with a sterile drape. Most centers
today use single-use pre-packed sterile drapes. These drapes have openings conveniently
placed at the location of patient's left and right inguinal area (Fig.5, Fig. 6).

Fig. 4. The table set for coronary angiography


The patient must be warned not to move his/her hands downward, towards the disinfected
areas of planned vascular access. The radiation shield and image intensifier/digital flat
panel detector are also covered with sterile covers. The manifold is connected and flushed,
and then zeroed for pressure monitoring.
Procedural Techniques of Coronary Angiography 99

Fig. 5. Sterile drape openings in patient's inguinal area, femoral pulse palpation

Fig. 6. Sterile drape openings in patient's inguinal area


100 Advances in the Diagnosis of Coronary Atherosclerosis

4. Catheter menu
Coronary angiography is performed with the use of specifically designed diagnostic
catheters. More than 95% of all procedures can be completed using catheters Judkins left 4
(JL4), and Judkins right 4 (JR4). Also, a pigtail catheter is used for obtaining left
ventriculogram, if it is recorded. Usually the catheters come pre-packed in a sterile
envelope, in which JL4, JR4 and pigtail catheters are present. Diagnostic catheters are more
elastic than the catheters used for PCI, and can be manipulated into the coronary ostium.
However, sometimes the anatomy of the aortic arch and coronary ostia requires the use of
different types of catheters. Today, the catheter menu is abundant with different versions,
and at least one of the available catheters will be able to adequately cannulate even the most
difficult and distorted coronary ostia (Fig.7). It is necessary to know the shapes of those
catheters in order to choose the appropriate-ones for certain anatomical variants.
Therapeutic (guiding) catheters are somewhat stiffer and do not forgive an imperfect
position, because the insertion of interventional equipment (coronary guidewires,
angioplasty balloons, stents etc.) creates much more backforce than does the radiocontrast
injection. Therefore, an unstable catheter with inadequate backup will be frequently
displaced out of the coronary ostium. This is why coronary stenting, even more than
diagnostic coronary angiography, requires the use of dedicated guiding catheters.

5. Arterial puncture
The standard technique of arterial puncture used today is Seldinger technique (Powell &
Moxey, 2011), cleverly developped by Sven-Ivar Seldinger, a Swedish radiologist, in 1953.
The procedure of arterial puncture is performed as follows: firstly, at the site of arterial
puncture, an appropriate pulse has to be palpated in order to locate the artery (Fig.5). A
local anesthesia is applied, usually with 10-15 ml of 1% or 2% lidocaine for local infiltration
of the skin and subcutaneous tissues. Then, a percutaneous puncture of the desired vessel
with a hollow beveled tapered-tip needle is made. The pointy tip at the distal end of the
needle is the first one in contact with the skin, and subsequently with the artery, while the
distal needle opening faces upwards. The needle is held in the operator's hand like a pencil,
at an angle of 30º-45º against the surface of the skin. Arterial puncture is made by advancing
the needle to the site of the arterial pulse. An experienced operator will usually feel when
the needle has punctured the arterial wall. A correct arterial puncure is confirmed by the
pulsatile back-flow of the bright-red blood from the proximal end of the needle (Fig.8).
It is not necessary to have a syringe attached to the needle during arterial puncture, because
the arterial pressure will produce a pulsatile jet of blood when the needle tip reaches the
arterial lumen. If the backflow is slow and barely pulsatile (or seems to be continuous,
without pulsations), and the blood is dark-red, the needle tip has punctured the vein. In
certain circumstances, however, even correct arterial puncture may resemble venous
puncture: shock, hypotension, severe bradycardia, very low oxygen saturation, cardiac
arrest and/or resuscitation etc.
After the arterial puncture, an atraumatic, curved-tipped guide wire (J-wire with moveable
inner core) is passed through the needle and advanced into the artery (Fig.9). The needle is
then removed, leaving the guide wire in the vessel. The site of puncture at the skin level is
usually widened by a couple of milimetres using a small and shallow cut by a surgical
scalpel, an 11-mm or 15-mm blade to facilitate the passage of the arterial sheath. The sheath
Procedural Techniques of Coronary Angiography 101

with a plastic dilator inside of it is advanced over the guide wire, and into the artery. The
dilator is removed, leaving the sheath in the vessel, and a haemostatic valve prevents
bleeding through the sheath. It also provides access for the passage of catheters and other
devices into the vessels, with no blood loss. The side arm of the sheath is aspirated and
injected with heparinized saline before the advancement of catheters. During arterial
puncture and sheath placement, the patient may develop a vasovagal reaction due to
puncture and vessel manipulation, which will manifest as hypotension and bradycardia.
Atropine IV usually swiftly reverts the condition, and sometimes elevation of patient's legs
and high-rate saline infusion with oxygen supplementation should be implemented
additionally. The technique of direct arterial approach, with the preparation and direct
visualization of the artery at the puncture site is today very rarely used, and therefore
discussed elsewhere.

Fig. 7. Diagnostic catheter curves, picture courtesy of Medtronic


102 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 8. Pulsatile back-flow of the bright-red blood from the proximal end of the needle

Fig. 9. J-wire is passed through the needle and advanced into the artery
Procedural Techniques of Coronary Angiography 103

6. Vascular access
6.1 Femoral approach
Preferences of particular access sites vary in different centers, but the most commonly used
are right femoral and right radial artery. The percutaneous transfemoral approach has been
routinely used for cardiac catheterization and coronary angioplasty (Kwac et al, 2010). Right
transfemoral approach is favoured because of the larger artery diameter, and therefore the
possibility to insert larger arterial sheaths, catheters and bulkier devices, in comparison with
the transradial approach. If there is a peripheral vascular problem, the left femoral, or right
or left radial or brachial approaches can be used. The femoral approach is the most
comfortable access for the operator. Its main disadvantage is the time-delay after the
angiography and before mobilizing the patient. After the femoral pulse is palpated (Fig.5)
and local anesthesia applied, the puncture of the femoral artery is performed some 2 cm
below the inguinal ligament (Fig.10), at the punctum maximum of the pulse. The inguinal
ligament is attached to spina illiaca anterior superior on one end, and with the pubic bone on
the other (Fig.10). This makes determining of its anatomical location easier, because
overlapping skin crease in obese patients can be located more distally, thus deceptively
making the less experienced operators prone to puncturing too distally. Puncture through
the ligament itself will make the haemostasis very difficult, because the ligament is a firm
structure which precludes adequate pressure application against the artery after
angiography.

Fig. 10. Inguinal area anatomy; 1 – spina iliaca anterior superior, 2 – inguinal ligament, 3 –
skin crease, 4 – common femoral artery, 5 – common femoral vein, 6 – femoral nerve, 7 –
deep femoral artery, 8 – saphenous vein, 9 – pubic bone, 10 – superficial femoral artery, 11 –
sartorius muscle, 12 – abductor longus muscle
104 Advances in the Diagnosis of Coronary Atherosclerosis

6.2 Radial approach


Alternative access when femoral puncture is unavailable or cannot be used, is radial or
brachial approach. While the femoral access is still most widely used in the USA and the
most of Europe, Asia and Americas, radial access is becoming more and more popular in
Europe, thanks to its advantage of avoiding prolonged periods of patients' bed-rest after the
procedure. The transradial approach has gained popularity in recent years mainly because
of the significantly decreased access-site complications and the greater patient comfort it
offers (Tomassini et al, 2011). However, the prevalence of transradial approach for coronary
procedures worldwide is still quite low and nowadays, in the United States, for example, the
favourite strategy for bleeding prevention is mostly based on the adoption of new
antithrombotic drugs (such as bivalirudin and fondaparinux) rather than on the selection of
an alternative, safer vascular access route (Rigattieri et al, 2010). Also, radial approach is
somewhat more difficult to master, and requires more training than the femoral one. The
main setback is the small diameter of the vessel, and the fact that the radial artery is prone to
spasm. Allen's test is also mandatory to determine the ability of the ulnar artery to take over
the circulation to the hand in case of an irreversible damage to the radial artery. Another
disadvantage of right radial approach is the considerable technical difficulty in evaluating
left interior mammary arterial graft. Instead, the left radial approach can be used. The third
reason because of which radial artery might not be used is the inability to use that particular
artery as a potential free arterial bypass-graft, should the bypass-surgery be indicated. Also,
there has been some controversy in radiation dose to the operator when using radial
approach. There were reports of significantly higher radiation doses for the operator in
comparison to femoral approach, especially during the learning phase. Before proceeding
with transradial coronary angiography, it is wise to administer premedication in order to
reduce the probability of radial artery spasm or thrombosis. Premedication cocktail
containing nitroglycerin and verapamil has been documented to be effective in preventing
radial spasms. However, verapamil is relatively contraindicated for some patients with left
ventricular dysfunction, hypotension and bradycardia. Intra-arterial premedication with 100
μg of nitroglycerin and 3,000 units of heparin is effective in preventing radial spasms during
transradial cardiac catheterization, according to Chen et al (2006). Some authors recommend
an intra-arterial cocktail consisting of 5 mg of verapamil plus 200 µg nitroglycerine in 10 ml
of normal saline, and others (Pancholy et al, 2006) use 200 µg/ml of nitroglycerin solution
injected on the lateral and medial aspect of the previous location of the radial artery
pulsation, should the arterial spasm occur after administration of lidocaine for local
anesthesia, and before radial cannulation. During transradial coronary angiography, the
wire should always go smoothly. If the operator feels some resistance, a peripheral radial
angiogram should be taken. There is a possibility that a radial loop with recurrent radial
artery exists, and the operator will be forced to change the access site. Left radial approach
can be used comfortably: it is possible to bend the patient's arm over the abdomen and
perform the procedure. Left radial approach is also favoured when re-evaluating left interior
mammary artery (LIMA) to left anterior descending coronary artery (LAD) bypass grafts.
The transradial approach can be safely proposed for all patients, with a low conversion rate
to transfemoral approach when an attempt on both radial arteries is considered first
(Guédès et al, 2010). Instead of JL4 used in transfemoral approach, for an average sized
patient, a JL3.5 can be used. The catheters should always be exchanged over a long wire, so
Procedural Techniques of Coronary Angiography 105

that renegotiating the wire through this spasm-prone and often tortuous vessel can be
avoided. Some population studies have shown that patients prefer radial access because of
early ambulation (Cooper et al, 1999) and some cardiologists also strongly prefer it.
However, one should bear in mind that the most important indication for PCI today is acute
myocardial infarction. This important setting must be considered when discussing the
preffered vascular access, because this is the primary circumstance in which PCI improves
survival. It can be expected that difficulties with radial approach and conversion to femoral
approach may delay time to reperfusion, jeoppardizing the lives of critically ill patients,
especially those in cardiogenic shock, requiring intra-aortic balloon pump, for example. So
the question is: with the increased use of radial approach, should it become the new
standard of care, especially regarding emergency interventions for acute myocardial
infarction, and can primary PCI (pPCI) be routinely performed through radial
approach?And the answer is: No. As for the setting of acute myocardial infarction (AMI),
Vorobcsuk et al (2009) performed a systematic review and meta-analysis of 12 trials
comparing the radial versus the femoral approach. They found no significant difference in
the time to reperfusion between the two groups but found access site failure when
transradial approach was used, which contributed to longer procedure times (Jolly et al,
2009). Recent systematic reviews and meta-analyses of trials in the acute setting of ST-
elevation AMI, where time is of the essence, have shown radial access to be associated with
a 10% crossover rate increasing "time to reperfusion" in the individual case (Vorobcsuk et al,
2009). Those trials usually excluded unstable patients in cardiogenic shock requiring
additional treatment options, such as intra-aortic balloon pumps, central venous access or
temporary pacing. These patients are automatically treated using femoral access as it is
more convenient and appropriate to use one access site. In some circumstances these
patients arrest on the table necessitating cardiopulmonary resuscitation, and femoral access
is simply easier and less fiddly under difficult circumstances (Patterson & Foale, 2011).
Radial artery puncture can only be attempted once. If that attempt fails, the morbidity
associated with access site crossover occurs, including the risk of radial artery occlusion, but
also the need for double site post-procedural care (Stella et al, 1997). The radial approach
therefore increases procedure times, not only because of the steep learning curve to this
technically more challenging approach. Radial artery anomalies are also relatively common
and are a cause of transradial procedure failure even for experienced radial operators (Lo et
al, 2009). Even when operators achieve equal procedural times using radial and femoral
approach, there is still significantly greater fluoroscopy time with radial approach PCI.
Inevitably an increase in the irradiation of staff and patient follows (Brasselet et al, 2008).
Femoral access is easier for training, not limited by anomalous radial artery anatomy, and
allows for both left and right mammary arterial bypass graft evaluation. It is also convenient
if a venous puncture is necessary for obtaining haemodynamic measurements. In these
circumstances, an adjecent femoral vein puncture requires "same-site" postprocedural
management. Surely this does not mean that the transradial technique should be dismissed
as a cardiology fashion. It should be learned because it has certain advantages in diagnostic
coronary angiography and elective PCI, it is associated with an earlier patient discharge, and
can be applied in circumstances of severe aortoiliac disease which precludes the use of
femoral approach. However, in primary PCI, the current enthusiasm for using transradial
approach is not sufficiently justified in evidence, and especially not justified enough to
dismiss the femoral route from current training programmes.
106 Advances in the Diagnosis of Coronary Atherosclerosis

6.3 Brachial approach


Brachial approach remains an option too, most commonly the right brachial approach. The
brachial artery is the distal continuation of the axillary artery, and itself separates distally
into ulnar and radial arteries. Percutaneous transbrachial approach is useful and safe
alternative for performing coronary angiography as well as PCI. The advantage of the
brachial approach as an alternative to the radial one is that brachial approach needs no
special premedication, and can be performed using the exactly same equipment (arterial
sheath and catheters), often even the larger-ones, including 7 Fr size, although most
operators usually use the shorter brachial sheaths. For brachial approach it is necessary that
an arm-board be placed for the patient's arm and hand during the preparation of the patient
for the procedure. The antecubital fossa should be prepped and draped, and the technique
of local anesthesia, arterial puncture, sheath placement etc. is generally similar to the one
used for femoral access. When puncturing the brachial artery, care must be taken not to
puncture the posterior arterial wall too. The complication rate is relatively low, and in the
large series has not exceeded 2% (Gonzalez et al, 1991). Complications of this site of arterial
access include: brachial artery thrombosis, hemmorhage, pseudoaneurysm formation,
cellulitis, phlebitis, and median nerve traction with possible irreversible damage.

6.4 Axillar approach


Axillary artery cannulation is a useful technique for obtaining central arterial access in
patients with severe peripheral vascular disease in the lower and upper extremities,
although it is very rarely used today. The axillary artery, which is the direct continuation of
the subclavian artery, courses from the outer margin of the first rib to the tendon of musculus
teres major, where it becomes brachial artery. The lateral portion of the axillary artery is
cannulated, just lateral to the head of musculus pectoralis minor. The patient's underarm must
be shaved and prepped in sterile fashion. The arm is hyperabducted and externally rotated
in order to access the artery (Gonzalez et al, 1991). After applying local anesthesia, a skin
incision is made using a no. 11 blade, and through that incision, an 18-gauge needle is
advanced until pulsatile backflow is obtained. Care must be taken to only perform a single-
wall puncture. Then, a J-wire and thereafter a side-arm sheath-dilator are placed into the
artery as with other accesses, and angiography can be conducted. After the procedure, the
patient is advised not to use the arm for at least one day to avoid bleeding. Complications of
axillary arterial cannulation include arterial thrombosis, brachial plexus injury, hemorrhage,
and pseudoaneurysm formation.

7. Performing the coronary angiography


Through the sheath placed into peripheral artery through which the vascular access is
enabled, and over the guidewire, the first coronary diagnostic catheter is introduced
through the peripheral artery (Fig.11), and farther into the aorta. Care must be taken that the
atraumatic J-tip of the guidewire precedes the catheter, as to avoid injury to the vascular
endothelium. The J-wire is 0.035 or 0.038 inch in diameter. There are many different types of
guidewires, different in length, structure, coatings, diameter, tip structure and shape, and
materials, available for different diagnostic and interventional vascular procedures. The
usual choice is the 0.035 inch wire with a 3 mm J-tip. For tortuous vessels, a hydrophilic
wire, such as Terumo Glidewire, or Wholey Wire with soft, floppy tip may be used.
Glidewire should not be used through the needle, because of the possibility to damage the
Procedural Techniques of Coronary Angiography 107

hydrophilic coating. Also, when advancing the catheter, the proximal part of the J-wire has
to be visible on the outside of the catheter and secured by the operator's hand (Fig.11), as to
avoid embolization of the J-wire into the patient's circulation. That would require an
emergency vascular surgical procedure to remove the J-wire from the patient's aorta. Under
the X-ray guidance, the guidewire tip is advanced up to the ascending aorta, and into the
aortic root. Holding it in place, the coronary diagnostic catheter is advanced over the
guidewire until it reaches the ascending aorta.
After the catheter reaches the Valsalva sinus in the aorta, the guidewire is withdrawn
leaving the catheter in place. The outside portion of the catheter is connected to a manifold
(Fig.12). The manifold is on the other side connected to the syringe (or an automated
injection-device) for radiocontrast application, and on its side arms to the saline infusion, to
the transducer for invasive blood-pressure monitoring, and to a radiocontrast container
from which radiocontrast is taken into the syringe. Through the manifold, the operator
controls the contrast injection, blood pressure, injections of heparinized saline if necessary,
etc. To ensure that there is no air or clot within the coronary catheter, a syringe is connected
to the catheter to aspirate and discard at least some 5 ml of blood. This is the approximate
volume of the catheter lumen. Some operators instead just allow a small ''bleed back'' from
the catheter, after which the catheter is connected to the three-port manifold.

Fig. 11. Through the arterial sheath and over the guidewire, the first coronary diagnostic
catheter is introduced into the artery; note the proximal end of the wire outside of the
catheter
When connecting the catheter to the manifold, it is important to connect using the fluid-
against-fluid method, i.e. to let the blood back-bleed from the catheter, and at the same time
to allow saline flush from the manifold. This manner of connecting the catheter to the
manifold prevents air embolism from air bubbles from within the catheter, or from within
the manifold.
108 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 12. Manifold with connections

7.1 Left coronary artery engagement


Under fluoroscopic guidance, the catheter is advanced into the coronary ostium, and several
recordings of the left coronary artery are taken. The most commonly used catheter
diameters are 6 Fr, but diameters from 4 Fr to 8 Fr can also be used. Diagnostic and
therapeutic coronary catheters are pre-shaped to suite specific aortic and coronary anatomy.
The most commonly used diagnostic catheters for coronary angiography are JL4 (Judkins
left 4 cm curve) for the left, and JR4 for the right coronary artery. Both types of catheters
were ingeniously designed by Judkins, and are easy to use, because they can engage the
coronary ostia with very little manipulation. In fact, Judkins himself said that "no points are
earned for coronary catheterization – the catheters know where to go if not thwarted by the
operator" (Judkins, 1968). When cannulating the left coronary artery, the catheter is
supposed to be unfolded in en face position in the ascending aorta. The best view to see the
coronary ostia (both left and right) and to perform the ostial coronary cannulation is left
anterior oblique (LAO) 50°, because sinus of Valsalva and aortic root are not superimposed
on either of the coronary ostia. Cannulating the left coronary artery (LCA) in antero-
posterior (AP) position is not recommended because it is difficult to appreciate the ostium,
and ostial Left Main Coronary Artery (LMCA) stenoses can be easily missed.
For engagement of the LCA, the J-wire is advanced to the level of aortic valve. Then, the JL4
catheter is placed as low as possible facing the left coronary ostium. The guidewire is slowly
withdrawn, and the catheter tip is already in the LMCA or just below it. If it is below the
ostium, than it should be withdrawn ever so slightly, and it will fall into the LMCA ostium.
Some operators fear that this method might be a bit risky, so they prefer to leave the catheter
tip above the level of the left sinus of Valsalva, then withdraw the guidewire from the
Procedural Techniques of Coronary Angiography 109

catheter, and thereafter advance the catheter tip downwards and into the LMCA ostium.
This, on the other hand, carries some risk of scratching the surface of the aortic endothelium
and embolizing atheromas if they are located in catheter's path. If the catheter tip is at the
level of the ostium, but not within the ostium itself, it is usually easily inserted into the
ostium by a clockwise or counter-clockwise torque. An injection of 5-10 ml of contrast close
to the expected location of the coronary ostium reveals the position of the catheter tip within
the aortic root and facilitates a precise cannulation. If the aorta is dilated, a JL5 or JL6
catheters may be required, while in small individuals, or with small aortic arches, a JL3.5 or
even JL3 catheters may be used to cannulate the left coronary artery. Multiple recordings are
taken from diffrerent perspectives. The optimal positions for recording the left coronary
artery are discussed later in the chapter.

7.2 Right coronary artery engagement


After recording the luminograms of the left coronary artery, the catheter is removed over
the J-wire, and another catheter, the one for the right coronary artery (RCA) is inserted
through the sheath and into the aorta, up to the right coronary ostium, also in the LAO
position. After reaching the ascending aorta with the tip of the right coronary catheter and
removing the guidewire from inside of it, the catheter is aspirated and connected to
manifold. The JR4 catheter is advanced into the aortic root, some 2 cm above the valve
level. Then, the operator applies a clockwise rotation with some traction to the catheter.
The catheter is slowly rotated using the rotation swivel at the distal portion of the
manifold, where the coronary catheter is attached. As the catheter rotates, the torque
should be reduced, so that there is no overshoot of the catheter over the right coronary
ostium. The catheter rotates and descends somewhat, thereafter cannulating the right
coronary ostium. If the first rotation is unsuccessful in cannulating the artery, the catheter
should be rotated counter-clockwise to the initial position as to avoid the kinking of the
catheter. The problem to find the coronary artery is most commonly due to the origin of
the RCA superiorly and more leftwards than usual. It may even arise from the left
coronary sinus, close to the LMCA. Using an Amplatz left (AL) 1 or 2 coronary catheter
can be helpful to perform selective cannulation of such an RCA. Usually two, but
sometimes more right coronary artery luminograms from different projections are
recorded, and the catheter is removed. If the right coronary artery ostium is oriented
straight downwards, a multipurpose (MP) catheter may be required. In contrast, if the
ostium is oriented upwards, a "shepherd's crook" (SC) catheter or internal mammary (IM)
catheter may be required. The optimal positions for recording the right coronary artery
are discussed later in the chapter.

7.3 Contrast injection technique


The syringe with radiocontrast is held with the handle elevated, so that any air-bubbles
inside of the syringe can rise to plunger, and are less likely to be injected into the patient's
coronaries. A significant air-embolism into a coronary artery may result in asystole, and
cardiac massage should be initiated. This usually reverts pretty quickly, as the air
components of the bubble (nitrogen, oxygen, carbon-dioxide etc) are dissolved in the blood.
Of course, the operator must previously make sure that there are no air-bubbles inside of the
syringe. The radiocontrast must be injected vigorously enough to ensure that it temporarily
110 Advances in the Diagnosis of Coronary Atherosclerosis

replaces the blood in the coronaries, with a continuous back-flow into the aortic root. A
weak injection may appear as a pulsatile contrast flow with filling-defects, and fail to
display the coronary lumen adequately. It sometimes causes the operator to declare a
coronary stenosis existence in a place where only a simple contrast filling defect exists. On
the contrary, a too-forceful injection can, in extreme cases, cause a coronary dissection,
although this event is extremely rare today. If there is no contrast back-flow to the aortic
root, or if the back-flow is inadequate, there may be an ostial lesion present.

7.4 Pressure damping


During the cannulation of the coronary artery ostia, a special care must be taken to
monitor the blood pressure. A sudden fall in blood pressure may lead to ventricular
fibrillation and cardiac arrest. An immediate defibrillation usually restores the normal
heart rhythm, although sometimes a full spectre of cardiopulmonary resuscitation has to
take place, along with the cardiac massage, endotracheal intubation, artificial ventilation
etc. This usually occurs if there is a severe ostial stenosis in the LMCA or the RCA, or if
the artery is very prone to severe spasms, or if the conus branch is superselectively
cannulated during the RCA engagement. It is a sign of a total coronary obstruction.
Pressure damping or "ventricularization", called so because the pressure waveform
resembles that of the ventricle, especially with very low diastolic pressure, can also be the
result of placement of the catheter tip with the end-hole against the vessel wall, or
presence of a thrombus, or an air bubble inside the catheter lumen, and sometimes also
occurs after kinking the catheter body.
Whenever blood-pressure damping occurs during the coronary artery engagement, it is
best to remove the catheter immediately (Conti et al, 1980). A repeated, careful
cannulation may be attempted, best preceded by administration of nitroglycerin. If
pressure damping occurs again, the angiogram can be swiftly recorded, and the operator
must remove the catheter immediately. Sometimes, a non-selective or semi-selective
opacification from the aortic root, with the catheter just in front of the coronary ostium
may be sufficient to delineate the ostial coronary disease. There is also an option to use
the side-hole catheters, which enable coronary perfusion through the sideholes located
just proximally to the catheter tip.

8. Coronary anatomy and the purposeful angiographic projections


Since the coronary arteries are three-dimensional structures, and the X-ray image is two-
dimensional, several different perspectives of observing the coronary luminograms are
necessary in order to adequately depict the coronary artery anatomy, the location, the
extent, and the degree of coronary obstruction. The projections are defined according to the
position of the digital flat panel detector, or image intensifier, depending on what kind of
cardioangiograph is used. The X-ray tube is under the cardioangiograph table (Fig.1), i.e.
under the patient. The positions can be defined as a combination of antero-posterior (AP),
cranial, caudal, left or right lateral, and left or right anterior oblique (LAO and RAO,
respectively). Extremely rarely, practically almost never, are posterior oblique positions
used in diagnostic coronary angiography. The eleven regularly used positions at varying
angles are: AP, AP-cranial, AP-caudal, left lateral, right lateral, RAO (right anterior oblique),
Procedural Techniques of Coronary Angiography 111

RAO-cranial, RAO-caudal, LAO (left anterior oblique), LAO-cranial, and LAO-caudal. These
positions are also defined by angles to cranial or caudal, and angles to the left or to the right.
These angles may vary depending on patients' anatomy, coronary anatomy, position of the
heart etc.

8.1 Radiological coronary anatomy


Left coronary artery is a complex structure, and demands usually at least 4-5 projections. In
those, the Left Main (LM, or LMCA) coronary artery has to be clearly visible, along with its
bifurcation and branches. The LMCA is a short artery, with variable length, usually
bifurcating into the left anterior descending and left circumflex coronary artery. Sometimes
LMCA can be very short, almost non-existant as such, in which case there is a "shotgun-
ostium" of the left anterior descending (LAD) and the left circumflex (CX) coronary artery.
The LMCA is rarely longer than 2 cm.
The LAD passes down the anterior interventricular sulcus and usually extends around the
heart's apex. It usually gives rise to two diagonal branches (D-1 and D-2), although they
may also vary in number. Diagonal branches are positioned on the left anterolateral part of
the heart's surface. The LAD also gives rise to septal perforators. The first one (1st septal) is
usually the largest of all the perforators. It marks the border between the proximal and the
mid portion of the LAD. The distal portion of the LAD is the distal 1/3 of the vessel, and has
no specific landmark as to define it against the mid part. It is important to note the largest
septal branch if the patient is to be subjected to alcohol septal ablation for hypertrophic
obstructive cardiomyopathy.
The circumflex artery is positioned in the atrio-ventricular (AV) groove and is located on the
left side of the heart. It gives rise to several atrial branches, but also to very important
marginal branches. As they are positioned on the margo obtusus cordis, they are called the
obtuse marginal (OM) branches. There are usually three of them (OM-1, OM-2, and OM-3),
but their number can also vary individually. Marginal branches are important to define,
because they are an available target for coronary bypassing if surgical revascularization is
planned. The CX artery is usually anatomically non-dominant, meaning that it does not give
rise to posterior lateral (PLA), nor posterior-descending (PDA) artery, which in that case
originate from the right coronary artery (RCA). If the CX continues through the AV-groove
and gives rise to PLA, while the PDA originates from RCA, than CX and RCA are co-
dominant. If the CX gives rise to both PLA and PDA, while the RCA is small and terminates
before bifurcating to PLA and PDA, than the CX is anatomically dominant. In this case, it is
usually the largest artery in coronary circulation. If it is non-dominant, than the territories of
blood supply of both LAD and RCA are larger and more significant than the CX one.
The right coronary artery (RCA) can vary in the position of its ostium. It gives rise to conus-
branch, which supplies the right ventricular infundibulum. The second most proximal
branch of the RCA is the sinoatrial node artery. The RCA continues on along the right-side
AV-sulcus giving rise to a few right-atrial branches. It also gives rise to one or two acute
marginal branches, called so because they are positioned on the margo acutus cordis. They
supply the right ventricular myocardium. Before reaching the crux cordis, the RCA also
gives off a few posterior right ventricular branches on the right-diaphragmatic surface of the
heart. Finally, at the crux cordis, on the heart's posterior wall, the RCA is bifurcating into the
PLA and the PDA. This RCA dominance is the most common situation, present in some 85%
112 Advances in the Diagnosis of Coronary Atherosclerosis

of all human hearts. The PLA and the PDA have several smaller branches, e.g. the posterior
septal perforators originating from the PDA. In the crux region, the atrioventricular node
artery origins from the RCA, and passes vertically upward towards the AV-node. This is
why ischemia in the region of RCA at the time of an acute inferior-wall myocardial
infarction can be associated with different degrees of heart block.

8.2 The best projections for evaluating the left coronary artery
Left caudal view (LAO-caudal, or "spider" view) is excellent for evaluation of the LMCA
and its bifurcation into the LAD and CX, or trifurcation into the LAD, CX and intermediate
branch if it exists (Di Mario and Sutaria, 2005). Interventionalists use it in many cases for
wiring the LAD or the CX. It forshortens the mid and the distal segment of the LAD, and
should not be used to evaluate these segments, although it can pretty nicely separate LAD
from diagonals and septals. Also, it can be very useful in viewing ostia of high marginal
branches, and therefore is used for wiring these branches too. It is usually done in 25-40º
caudal and 30-50º to the left.
Left cranial (LAO-cranial) view is very good for showing the ostium of the LMCA, but
considerably forshortens the proximal half of the LAD. It is convenient for separating the
diagonals and the septals from the LAD, though, and can be used to guide the guidewire
into and from these branches during interventions. The CX and its high-originating
branches are not appreciated well in this projection, but if the CX is dominant, this
projection can be useful to depict the very distal parts of the CX.
Cranial (AP-cranial) projection displays the ostium of the LMCA (Di Mario and Sutaria,
2005), similarly to the LAO-cranial view. Although some operators use it to intervene on the
proximal LAD, the aouthor's opinion is that the overlap of the CX is too distracting, and that
there are better projections for interventions and evaluation of the proximal LAD than the
AP-cranial. This projection can be useful for evaluating the mid and distal segments of the
LAD and diagonals' take-off, although the diagonal ostia can be overlapped with the main-
course LAD. The CX in this view often looks confusing for the inexperienced operator,
especially in its proximal and mid segments, but if it is dominant, this can be a very useful
projection for evaluating the very distal portions of the CX.
Caudal (AP-caudal) is a very good projection for evaluation of the LMCA, its bifurcation (or
in some cases trifurcation), ostial LAD, and the intire CX if it is not dominant. This is the
author's favourite projection for interventions on the CX. It can be useful for ostial stent
placement in the LAD, similarly to LAO-caudal. Some operators prefere wiring of the
marginal branches in RAO-caudal, but in that projection, the marginal ostia are often
overlapped with the main course CX. The author feels that AP-caudal is the best projection
for evaluating the CX in its entirety, including the marginal ostia, meaning that this
projection is also probably the best for wiring those too. It is especially good for treating
bifurcational lesions on CX + marginals, but in author's experience also good for treating
bifurcation lesions on LMCA + LAD + CX, along with the spider-view. However, if the CX
is anatomically dominant, the distal portions of the artery will usually overlap with 3rd
marginal branch, PLA and/or PDA. For these, in the case of CX dominance, the cranial
projections should be used.
Right cranial (RAO-cranial) projection is the most useful if it is performed with increased
cranial angulation of 30º, or in some cases even more. This projection shows nicely the mid
Procedural Techniques of Coronary Angiography 113

part of the LAD with the take-offs of diagonal branches and can in most instances be very
useful for treating bifurcation lesions within the LAD + diagonals. Sometimes, the diagonal
ostia will overlap, and LAO-cranial, LAO-caudal or AP-cranial will be better for that
purpose. If the LMCA is very short, even proximal and ostial LAD can be nicely appreciated
in steep RAO-cranial. The CX can be appreciated in this view only if it is dominant, and that
goes only for its most distal parts. The proximal and the mid segment of the CX cannot be
adequately evaluated in this view.
Right caudal (RAO-caudal) view is very good for evaluating the distal LAD. The proximal
and the mid part of the LAD are overlapped with diagonals in most cases, so this should be
taken into consideration when evaluating LAD in RAO-caudal. Still, some stenoses of the
LAD can only be seen here, so it requires operator's attention. The LMCA is displayed in
this view, although the bifurcation itself is being overlapped in most patients by the ostial
segments of the LAD and the CX. The CX artery can be nicely evaluated in RAO-caudal, but
to be sure, the AP-caudal should always be taken.
Left lateral is rarely used today. It can be useful to display LIMA-LAD anastomosis, the mid
and the distal portions of the LAD, and to separate the LAD from the diagonals, LAD being
the closest to the sternum. Pure RAO is rarely used today, since it does not display any of
the LCA segments ideally, except it can be useful for evaluating the distal LAD. Pure LAO is
very useful for cannulation of both LCA and RCA, but is not very useful in evaluating LCA
or its branches, so it is not being routinely used for LCA today. Straight AP is also not an
ideal projection for evaluation of the LCA, and for any of the segments, there are many more
useful projections.

8.3 The best projections for evaluating the right coronary artery
Many operators believe that two projections can be quite enough for evaluation of the RCA
since it has less complex anatomy in comparison to the LCA. The RCA can be viewed nicely
in LAO, but even better in LAO-caudal, which is better than the former one in displaying
the crux. The crux with the bifurcation of the RCA into the PLA and the PDA is also very
nicely displayed in AP-cranial, and in some cases in RAO-cranial projection. Some operators
use only simple LAO and RAO to evaluate the RCA. Left cranial (LAO-cranial) displays best
the proximal and the distal RCA and is very good for displaying the crux. It can also be
pretty good for displaying the PLA and the PDA (Di Mario and Sutaria, 2005). Cranial (AP-
cranial), sometimes with just a small angulation to the right (5-10º) is very good for
displaying the crux, the PLA and the PDA. The mid-RCA is best displayed in lateral and the
RAO-view, but in most cases, LAO can be sufficient for this segment also.

8.4 Angiographic studies and engagement of coronary bypass grafts


When a previously operated patient, who had undergone a bypass-coronary
revascularization, is subjected to coronary angiography, beside evaluating the native
coronary arteries, a study of bypass-grafts is also necessary. The operator should read the
surgical report in order to find out how many, and which bypass grafts there are. The
venous, or free arterial bypass grafts, i.e. the bypass grafts for aorto-coronary bypassing are
best cannulated from RAO 40°, but some operators cannulate them from LAO 50°, just as
they do for native coronaries. Still, from RAO 40° there is the best depiction of the grafts'
origin from the aorta. The bypass graft for the RCA can usually be cannulated using the
114 Advances in the Diagnosis of Coronary Atherosclerosis

standard JR4, or JR3.5, sometimes JR3 catheter. The origin of the graft to the RCA is usually
in the ascending aorta, just above the sinotubular junction. The cannulation is performed by
placing the catheter high in the ascending aorta, applying clockwise-rotation, and, at the
same time, pushing the catheter a bit downwards. If Judkins catheters are inadequate, RCB
(right coronary bypass) catheter can also be used for cannulating the SVG (saphenous vein
graft) to RCA.
The JR4 or JR3.5 catheters can also be used to cannulate all the other bypass grafts. Those
grafts' origins are usually located higher in the ascending aorta, just beneath the arch. If the
grafts for LAD, diagonals, intermediate, and obtuse marginals are unreachable with JR
catheter, an LCB (left coronary bypass), or an AL catheter can be used. If grafts cannot be
found selectively, an aortography in LAO position can be used to determine their origin,
position, or stumps if they are occluded. For obtaining a proper evaluation, after
aortography, a selective cannulation of the grafts should be reattempted.
Left internal mammary arterial graft can be cannulated using the IMA (internal mammary
artery) catheter, but JR4, JR3.5, JR3, or short tip JR can also be used in certain situations. The
ostium of the left internal mammary artery (LIMA) is engaged in 50° LAO position,
although sometimes it can be viewed better from AP or RAO 40° position. First in 50° LAO,
a J-wire is passed into the left subclavian artery, beyond LIMA. Than the IMA catheter is
passed over the wire, also beyond the LIMA origin. Then, from the position best suited for
the operator (AP, LAO 50°, or RAO 40°), the catheter is slowly withdrawn, rotating the tip
counterclockwise. Small test-injections are needed to display the ostium of the LIMA. The
angiography should be performed from several positions, especially paying attention to the
distal anastomosis with the LAD, since this position is the one most prone to iatrogenic
injury from manipulation during the grafting procedure, thus being the most prone to
developing a significant stenosis. A 90° left-lateral position is almost mandatory in
displaying LIMA-LAD graft, since it is very good to depict the distal anastomosis.
For cannulating the right interior mammary artery (RIMA), the brachicephalic artery is
engaged, J-wire is advanced distally to the RIMA origin, from the right subclavian to the
right axillary artery. The IMA catheter is advanced also distally over the wire. In the LAO
50° projection, after removing the J-wire, the catheter is slowly withdrawn, and with
occasional test-injections engaged into the RIMA ostium.
The right gastroepiploic artery has been used for coronary bypass, although very rarely.
Surgeons sometimes use it when mammary arteries and saphenous vein grafts are
unavailable. It is cannulated by first entering the hepatic artery using the cobra catheter. The
operator must then advance a hidrophylic guidewire into the gastroduodenal artery, and
then into the right gastroepiploic artery. Selective angiography of the letter will be enabled
by switching the catheter, usually to right Judkins (Di Mario and Sutaria, 2005). The
procedure is performed very rarely, and because of the unusual anatomy for the invasive
cardiolgist, can be time-consuming.

9. After the procedure


The operator should always review his/her angiographic recordings before withdrawing
the catheter, because standard projections used may not adequately reveal the condition of
the coronary circulation, and additional shots may be necessary. The further therapeutic
Procedural Techniques of Coronary Angiography 115

strategy should be considered (medical, surgical or interventional treatment). After the


catheter is removed, and the procedure is finished, the patient is transfered out of the
catheterization laboratory. Arterial sheath is removed, and manual pressure is applied onto
the site of arterial puncture in order to stop the bleeding. That is followed by mechanical
compression of that site in order to definitely stop the bleeding. The patient is later
mobilized, and if there is no indication for the patient to stay in the hospital further, he/she
is discharged on the next day, or sometimes on the same day after the procedure.

9.1 Haemostasis
Haemostasis is an important practical matter. Complications from diagnostic cardiac
catheterization are rare, but if they happen, those related to haemostasis are the most
common ones. When a patient comes out of the hospital after coronary angiography or
coronary stenting, and gets a groin haematoma, he/she will most likely quickly forget the
top-notch, several million euros costing equipment used during the procedure, as well as
the skills of the operator who performed sometimes very difficult intervention, but will
remember a large haematoma, and often complain about it to his family, neighbours, and
friends. Therefore, it is very important to adequately perform haemostasis, not only for
obvious medical, but also for "public-relation" reasons. The details of haemostasis and its
possible complications are discussed elsewhere. However, there are a few points that need
to be noted. Haemostasis may be difficult if the patient is obese, if there is high blood
pressure (always try to regulate the blood pressure prior to sheath removal), or if clotting
time is prolonged. If there is no vital indication, coronary angiography should be avoided if
INR is >1.6, otherwise, the sheath should be sutured and removed upon achieving a more
appropriate INR value. A significant aortic regurgitation with pulsus celer et altus can also
make the post-sheath-removal hold difficult.
Large sheaths create bigger holes in the arterial wall, so the bigger the sheath diameter, the
more difficult haemostasis will be. The patients who frequently elevate intra-abdominal
pressure by sneezing, coughing or performing Valsalva maneuvers can prolong bleeding
from the vascular access site. Same is true for restless patients, so those sometimes need to
be sedated and observed by a nurse. In order to facilitate the haemostasis process,
haemostatic devices have been developed, such as: collagen plugs, external pressure
devices, vascular clips, external C clamps, topical haemostatic accelerators etc.

10. Non-coronary left heart catheterization


Sometimes, an additional catheter, called pigtail-catheter (because of its shape) is inserted
into ascending aorta to perform aortography, and/or into the left ventricle to perform left
ventriculography. Today, with skilled echocardiographers and top-notch cardiac ultrasound
devices, these additional procedures are rarely used, but will be described too, so as to give
the readers the necesary technical information about the value of these procedures, as well
as how they should be performed.

10.1 Left ventriculography


Today, with skilled echocardiographers and excellent cardiac ultrasound equipment, there
is far less need to perform left ventriculoghaphy than previously. With reliable
116 Advances in the Diagnosis of Coronary Atherosclerosis

echocardiograms, avoiding left ventriculography spares the patient from additional contrast
administration, which is especially important if the patient already has impaired renal
function. Also, without left ventriculography there is less radiation both for the patient and
for the operator. If the patient is in heart failure or cardiogenic shock, every additional
volume intake can prove to be risky for his/her condition. However, some operators and
some centers prefer to perform the left ventriculography, especially if there is doubt about
echocardiography findings, or if there is no echocardiography finding available.
In order to perform the left ventriculoghaphy, the operator advances the pigtail catheter
over a J-wire to the aortic root. The author's choice is usually the angled version of the
pigtail catheter. The J-wire is withdrawn into the catheter, with the J-wire tip just inside the
catheter tip to add to the catheter stiffness in order to successfully pass over the aortic valve.
The position of the digital flat-panel detector or the image intensifier is in RAO 30°. The
catheter with the wire inside of it is advanced farther, so that the body of the catheter
"bounces off" the aortic valve, and the distal curve faces upwards. Then the catheter is
rotated in a clockwise fashion, being at the same time slowly withdrawn. This causes the
catheter to rotate, and when the aortic valve opens, slip through the aortic ostium and into
the left ventricle. If there are difficulties to pass the catheter into the left ventricle this way,
the catheter tip is withdrawn into the ascending aorta, and the valve is probed with the J-
wire. When the wire passes into the ventricular cavity, the catheter is advanced into the
ventricle over the wire. Sometimes, a Glidewire with hydrophyllic coating must be used to
pass the aortic valve, especially if there is a tight aortic stenosis. Also, sometimes it might be
easier to pass the valve with a Judkins right coronary catheter.
The catheter is then held with the tip in the middle of the ventricle. The proximal end of the
catheter is connected to the injector after it has been adequately aspirated and flushed.
Through the automated injector, some 35–50 ml of contrast is injected into the ventricle at
the rate of 12-15 ml/s, delineating the left ventricular cavity and displaying the left ventricle
contractility. Intracavitary thrombi can be seen as hypodensities or filling-defects of contrast
within the left ventricle. Mitral regurgitation, if exists, can be seen and semi-quantified,
using degrees from 1 to 4, with 4 being the most severe. Care must be taken when advancing
the pigtail catheter into the left ventricle, because the mechanical contact with endocardium
can provoke premature ventricular beats, even ventricular tachicardia. It can also provoke a
complete heart block if the patient already has a right bundle-branch block, and the catheter
irritates the left bundle-branch upon contact with left ventricular endocardium. If there are
foreign masses within the left ventricle, such as thrombi or vegetations, those can be
fragmented and embolized into systemic circulation, causing potentially catastrophic
outcomes. A combination of a fragile myocardium and too vigorous catheter manipulation
can cause myocardial rupture with fatal cardiac tamponade and refractory shock.
Entanglement of the catheter into the chordae tendineae of the mitral valve or hooking the
catheter tip on a mitral cusp can result in seemingly severe mitral regurgitation, so care
must be taken to avoid this. In exceptional circumstances, catheter tip can theoretically rip a
mitral cusp or break up a chorda or two, causing acute mitral regurgitation of various
degree, thus complicating the left ventriculography.
After the ventriculography, the catheter is connected to manifold to record pressures in the
left ventricle. Upon withdrawal of the catheter over the aortic valve, pressure curves in the
aorta are also noted, and pressure values recorded. The systolic pressures in the aorta and
Procedural Techniques of Coronary Angiography 117

the left ventricle are noted for transvalvular aortic pressure gradient. Care must be taken not
to declare aortic stenosis in patients with previously unrecognized hypertrophic obstructive
cardiomyopathy with high pressure-gradients across the left ventricular outflow tract,
which may mimic the haemodynamics of aortic stenosis, with pressure gradients between
the middle of the left ventricle and the ascending aorta.
Calculation of the left ventricular ejection fraction (LVEF) is usually performed using the
software already incorporated into the imaging review station. Some operators use the semi-
quantitative eye-ball method in LVEF estimation, which has, when used by experienced
operators, shown a surprisingly good correlation with the actual value of LVEF measured
invasively in several studies. The software calculates LVEF by subtracting left ventricular
end-systolic volume from the left ventricular end-diastolic volume.
Mastering the left ventriculography is important as the transfemoral – transcatheter aortic
valve implantation is being performed increasingly. For severely stenosed aortic valves, it
may be easier to pass the valve using an AL catheter and a Glidewire.

10.2 Aortography
Aortography is defined as a radiographic recording of injection of a large volume of
radiocontrast into the aortic root. Most commonly, some 50 ml of radiocontrast, at a rate
of 15-25 ml/sec with a 0.5 sec linear rise is used. The catheter of choice is a sidehole
catheter, usually the pigtail shape. The best projection for estimation of aortic
regurgitation is LAO 45°. Aortic regurgitation is semi-quantified and described in degrees
from 1 to 4, much like mitral regurgitation. If the patient is subjected to invasive
diagnostics because of suspected coarctation of aorta, the coarctation will be best visible if
aortography is performed in LAO 20° position. Using the pressure monitoring, the
gradient across the coarctation can also be measured. For evaluation of a suspected patent
ductus arteriosus, the shot is usually taken at a very steep LAO projection. Aortography is
usually done for evaluating the ascending aorta for dilation or aneurysm, and/or aortic
regurgitation. The J-wire and the catheter are positioned in the ascending aorta, inside the
root or at the level of sinotubular junction. Anatomy and dimensions of the ascending
aorta and aortic arch are noted, as well as aortic regurgitation (if such a disorder exists),
and/or aorto-coronary bypass grafts, also if they exist. Care must be taken that the
catheter does not protrude back into the left ventricle during injection, which can make
quantification of the aortic regurgitation difficult.

11. Conclusion
Invasive cardiologists today generally continue their education and become
interventionalists. Anyone who wants to become a good interventionalist must be able to
routinely perform diagnostic coronary angiography and to be able to solve the problems
that might occur during the procedure. Therefore, the techniques of performing coronary
angiography must be mastered by an invasive cardiologist. That creates the conditions for
swift procedures, informative findings, purposefull conclusions and further
recommendations, and last, but not least, the maximum safety for the patient. However,
even with most carefully conducted coronary angiograms, there can be false readings. This
is especially true for less experienced operators, who can sometimes misinterprete the
118 Advances in the Diagnosis of Coronary Atherosclerosis

finding, thus affecting the final recommendation for further treatment. These pitfalls may
be: inadequate number of projections, ostial coronary occlusions without visible stumps,
superselective injection, e.g. especially if LAD and CX have (almost) separate ostia, catheter
induced coronary spasm, congenital coronary anomalies, pulsatile injection of radiocontrast
with filling defects mimicking coronary atherosclerotic obstruction, myocardial bridging –
the key to recognize it is the fact that the lumen normalizes upon muscle relaxation, i.e.
during diastole. Today, coronary angiogram projections must be observed by the operator in
the context of future percutaneous coronary intervention. The lesions are not assessed only
as significant or non-significant, but are evaluated regarding their length, their position
regarding side-branches and other caracteristics important for the possible intervention.
Therefore, it is of paramount importance to adequately display the coronary arteries and
their lesions, in appropriate projections, with an adequate number of different views, still
remaining rational regarding radiation and contrast medium consumption. Interventional
procedures in cardiology today have expanded beyond the treatment of coronary artery
disease. We are witnessing a technopharmacological improvement which enabled the
interventional, catheter-based treatment of valvular heart disease, closure of interatrial and
interventricular septal defects, relief of aortic coarctations, interventional treatment of
hypertrophic obstructive cardiomyopathies, paravalvular leak closure, left atrial appendage
closure, peripheral vascular disease treatment etc, and the basis for performing any of the
interventional procedures, especially percutaneous coronary interventions, is mastering
coronary angiography first.

12. References
Brasselet C, Blanpain T, Tassan-Mangina S, Deschildre A, Duval S, Vitry F, Gaillot-Petit
N, Clément JP, Metz D. (2008) Comparison of operator radiation exposure with
optimized radiation protection devices during coronary angiograms and ad hoc
percutaneous coronary interventions by radial and femoral routes. Eur Heart J 2008
Jan;29(1), pp. 63-70, ISSN 0195-668x
Chen CW, Lin CL, Lin TK, Lin CD. (2006) A simple and effective regimen for prevention of
radial artery spasm during coronary catheterization. Cardiology. 2006;105(1): pp. 43-
7, ISSN 1878-5409
Conti CR, Levin DC, Grossman W. (1980). Coronary Angiography, In: Cardiac catheterization
and angiography, Grossman W, pp. 147-169, Lea & Febiger, ISBN 0-8121-0712-4,
Philadelphia, Pa, USA
Cooper CJ, El-Shiekh RA, Cohen DJ, Blaesing L, Burket MW, Basu A, Moore JA. (1999).
Effect of transradial access on quality of life and cost of cardiac catheterization: a
randomized comparison. Am Heart J 1999 Sep;138(3 Pt 1): pp. 430-6, ISSN 0002-
8703
Di Mario C & Sutaria N. (2995). Coronary angiography in the angioplasty era: projections
with a meaning. Heart 20052005;91: pp. 968-976, ISSN 1355-6037
Gonzalez A, Rahimtoola SH, Kulick DL. (1991). Technique of Vascular Access, In: Techniques
and Applications in Interventional Cardiology, Kulick DL and Rahimtoola SH, pp. 1-18,
MosbyYear Book, ISBN 0-8151-7048-3, St. Louis, MO, USA
Procedural Techniques of Coronary Angiography 119

Guédès A, Dangoisse V, Gabriel L, Jamart J, Chenu P, Marchandise B, Schroeder E. (2010)


Low rate of conversion to transfemoral approach when attempting both radial
arteries for coronary angiography and percutaneous coronary intervention: a study
of 1,826 consecutive procedures. J Invasive Cardiol. 2010 Sep;22(9): pp. 391-7, ISSN
1042-3931
Jolly SS, Amlani S, Hamon M, Yusuf S, Mehta SR. (2009). Radial versus femoral access for
coronary angiography or intervention and the impact on major bleeding and
ischemic events: a systematic review and meta-analysis of randomized trials. Am
Heart J 2009 Jan;157(1): pp. 132-40, ISSN 0002-8703
Judkins MP: Percutaneous transfemoral selective coronary angiography. Radiol Clin North
Am 6 1968 Dec;6(3): pp. 467-92, ISSN 0033-8389
Kwac MS, Yoon SJ, Oh SJ, Jeon DW, Kim DH, Yang JY. (2010). A rare case of radial
arteriovenous fistula after coronary angiography. Korean Circ J. 2010 Dec;40(12): pp.
677-9, ISSN 1738-5520
Lo TS, Nolan J, Fountzopoulos E, Behan M, Butler R, Hetherington SL, Vijayalakshmi K,
Rajagopal R, Fraser D, Zaman A, Hildick-Smith D. (2009). Radial artery anomaly
and its influence on transradial coronary procedural outcome. Heart 2009;95: pp.
410-415, ISSN 1355-6037
Pancholy SB, Coppola J, Patel T. (2006). Subcutaneous administration of nitroglycerin to
facilitate radial artery cannulation. Catheter Cardiovasc Interv. 2006 Sep;68(3): pp.
389-91, ISSN 1522-1946
Patterson T, Foale RA. (2011) If the radial artery is the new standard of care in primary
percutaneous coronary intervention, why is most intervention done by the femoral
approach? Heart 2011;97: pp. 521-522, ISSN 1355-6037
Powell D & Moxey CF. (2011). Diagnostic Catheterization, In: Invasive Cardiology: a manual
for cath lab personnel, 3rd Edition, Watson S & Gorski KA, pp. 143-162, Jones &
Bartlett Learning, ISBN 978-0-7637-6468-5, Sudbury, MA, USA.
Rigattieri S, Di Russo C, Silvestri P, Fedele S, Loschiavo P. (2010). Our technique for
transradial coronary angiography and interventions. Indian Heart J. 2010 May-
Jun;62(3): pp. 258-61. ISSN 0019-4832
Stella PR, Kiemeneij F, Laarman GJ, Odekerken D, Slagboom T, van der Wieken R. (1997).
Incidence and outcome of radial artery occlusion following transradial artery
coronary angioplasty. Cathet Cardiovasc Diagn 1997 Feb;40(2): pp. 156–8, ISSN 0098-
6569
Tomassini F, Gagnor A, Varbella F. (2011). Successful use of an extra-long hydrophilic-
coated sheath in enlarged aorta to overcome extreme tortuosity of right subclavian
artery via transradial approach during coronary angiography. J Invasive Cardiol.
2011 Mar;23(3): pp. 56-7, ISSN 1042-3931
Vorobcsuk A, Konyi A, Aradi D, Horváth IG, Ungi I, Louvard Y, Komócsi A. (2009)
Transradial versus transfemoral percutaneous coronary intervention in acute
myocardial infarction: systematic overview and meta-analysis. Am Heart J 2009
Nov;158(5): pp. 814-21, ISSN 0002-8703
120 Advances in the Diagnosis of Coronary Atherosclerosis

Watson S & Hunter J. (2011) Cath Lab Staff Duties, In: Invasive Cardiology: a manual for cath
lab personnel, 3rd Edition, Watson S & Gorski KA, pp. 119-129, Jones & Bartlett
Learning, ISBN 978-0-7637-6468-5, Sudbury, MA, USA.
7

Risks and Complications of Coronary


Angiography: Contrast Related Complications
S. Mohammad Reza Khatami
Nephrology Research Center, Tehran University of Medical Sciences
Iran

1. Introduction
Since Wilhelm Röntgen reported his novel discovery of the X- Ray in 1895(Röntgen, 1898 as
cited in Weinmann etal., 2005), the Diagnostic imaging has been an integral part of
medicine. He was decorated by the second class of Prussian crown medal and he was also
awarded the first physic Nobel Prize at 1901. Levy- Dorn was another pioneer in radiology
who understood the potential importance of X-ray in medicine and opened a daily private
office for medical examination with X-ray in 1896 just one year after innovation of X-ray by
Röntgen. He pointed to the danger of this method to the living organism very early
(Hierholzer K & Hierholzer J 1997; Nitze, 1895; as cited in Hierholzer K & Hierholzer J, 2002)
Since X-rays are not sufficiently absorbed by soft tissue, contrast media (CM) is used to
highlight organs or pathologies. Osborne etal reported that inorganic Iodine enhanced the
urinary tracts X-ray images (Osborn etal., 1923). But the most important contributor to the
field of urological imaging came from the field of agriculture when A Binz and C Räth, two
scientists from the Berlin agricultural college synthesized a new organic iodinated molecule
"Selectan"(Binz & Räth 1928 as cited in Hierholzer K & Hierholzer J, 2002), and a research
fellow used this agent in clinical investigations. (Swick 1929a, 1929b as cited in Hierholzer K
& Hierholzer J, 2002; Weinmann etal., 2005).
In 1928 an urologist, Forssmann, used one of these agents in his own coronary arteries
(Hricak & Barbarive 1984).This was an unexpected methodological contribution of
urological research to cardiology.
From 1929 on, serial production of Uroselectan was established. This agent was a big step
forward in safety, because ionic iodide (Sodium Iodide and Lithium Iodide) cannot be used
in a clinical setting. In the early 1950s, a new generation of iodinated agents was launched,
which were derivatives of triiodinated benzoic acid, e.g. diatrizoate (Urografin etc.). These
agents became standard in diagnostic imaging for the next 30 years and are still widely used
in many countries. Almen suggested that non-ionic molecules could reduce osmolality
(Almen, 1969, as cited in Weinmann etal., 2005), an idea that was the basis of safer
compounds that entered routine clinical use in the 1980s. Water-soluble, non-ionic iodinated
contrast agents are now the workhorses in contrast based imagings. More than 90% of
contrasts media are used in western countries are non-ionic CM.
There are two main complications of contrast media. The aim of this chapter is to discuss
these two major side effects of CM. the most important of these two complication is contrast
induced nephropathy (CIN) which will be described in details and the other is anaphylaxis
reactions to the CM which will be mentioned briefly at the end of the chapter.
122 Advances in the Diagnosis of Coronary Atherosclerosis

2. Contrast induced nephropathy


According to the report of American Heart Association statistics committee and stroke
statistics subcommittee updated at 2007, from 1979 to 2004 the number of cardiac
catheterization has been increased 334%. Meanwhile from 1987 to 2004 the number of
percutaneous coronary intervention (PCI) has also increased 326%.IN 2004 more than
1,297.000 cardiac catheterization and in 2006 at least 1,313,000 PCI were performed in U.S
(American Heart Association statistics committee and stroke subcommittee, 2007). In 2003,
about 80 million doses of CM were used worldwide (Persson, 2005). Now in U.S more tha 10
millions radiological examinations with CM are performed annually which 4 millions of
these procedures are cardiac catheterizations. In western countries 6000 diagnostic cardiac
catheterization and 2000 therapeutical procedure are done per 1million habitants (Costa,
2004; Ultramari etal., 2006). On the other hand the physicians are dealing with a significant
increase in the prevalence and incidence of cardiovascular diseases, chronic kidney diseases
and diabetes which are considered as risk factors of CIN development. All these patients are
in increased need to such a diagnostic and the therapeutical maneuver.
CIN is a very interesting Issue for researchers. It is a type of acute kidney injury (AKI) which
in contrast to all other causes of AKI, the exact time of injurious insult is known.
Accordingly, it may help the investigators to find a more preciously diagnostic tool beside
to develop more effective prophylactic and therapeutical interventions.
CIN is a matter of health conflicts. It is an important causes of increased resource utilization,
prolonged hospital stay, increased in hospital and long term mortality and provoke the
progression of CKD (Bartholomew etal., 2004; Goldenberg etal., 2009; Gruberg etal., 2000;
James etal., 2010; McCullough etal., 1997; Solomon etal., 2009; Subramanian etal., 2007).

2.1 Physicochemical properties and classification of Contrast Media (table1)


CM is derivatives of tri-Iodinated benzene and is classified according to their Ionization,
osmolality and structural properties. The ionized media are dissociated in water, while the
nonionic agents are not dissociated although they are soluble in water. The ratio of iodine to
dissolved particles, describes an important relationship between opacification and
osmotoxicity of a contrast agent, higher this ratio, more desirable. In high osmolal CM this
ratio is 1.5, which in low-osmolal and iso-osmolal CM this ratio is 3 and 6 respectively. With
lowering the osmolality, the viscosity will increase. Viscosity has some deleterious effect on
renal blood flow and urinary flow in tubules. Contrast agents are completely filtered. With
reabsorption of water in tubules the concentration of CM reaches 50-100 times of plasma
concentration.
The first generation CM was Ionic. They had 3 atoms of Iodine, one atom of sodium and 2
active osmotic particles in each molecule,therefore their osmolality was very high (1200-1800
mosm/kgH2o), so they called hyper-osmolal contrast media (HOCM). From late 80s, the
next generation of these agents has been marketed. They are non-Ionic. They have only one
somatically active particle. They have been known as Low-osmolal Contrast Media (LOCM)
because their osmolality is lower than first generation but yet significantly higher than
plasma osmolality (600—900 mosm/kgH2o). The third generation of CM is Iso-osmolal
Contrast Media (IOCM) (≈300 mosm/kgH2o). They are non-ionic dimers. They have two
benzene rings rather than one ring of previous generations. This structure allows 6 Iodine
atoms to attach to the one cosmetically active particle.
123
Risks and Complication of Coronary Angiography: Contrast Related Complications

amount of iodine
Type Name osmolality ionicity mono/dimer viscosity
iodine ratio
(mosm/kg water) mg/mL mPa.sec at 370c

HOCM Iothalamat 1695 Ionic monomer 400 3:2 2.8


LOCM Iomeprol 720 nonionic monomer 350 3:1 4.8
LOCM Iopentol 683 nonionic monomer 310 3:1 6.5
LOCM Iopromide 586 nonionic monomer 350 3:1 4.7
LOCM Iopamidol 653 nonionic monomer 300 3:1 4.6
LOCM Ioversol 719 nonionic monomer 320 3:1 6
LOCM Iohexol 667 nonionic monomer 300 3:1 5.7
LOCM Ioxaglate 584 ionic dimer 320 3:2 7.8
IOCM Iodixanol 290 nonionic dimer 320 6:1 11.4
Table 1. Types of contrast media and their physicochemical properties

2.2 Definition & diagnosis


Definition of CIN is a matter of controversy and there is no a unique accepted definition.
CIN is a rapid decline in renal function after CM administration while there is obviously no
other causes of acute kidney injury (AKI).
Serum creatinine is the standard marker for detecting CIN. Small changes in serum
creatinine after CM exposure are seen but it is not considered clinically relevant.
In last two decade, different criteria of renal function deterioration were defined in clinical
trials. But the two most popular are: 1- A 25% relative increase of serum creatinine from
baseline and 2- An absolute increase of 0.5 mg/dl of serum creatinine from the base. It will
happen within 2 days to one week after contrast exposure. None of these two criteria are
accurate. The most common method for the assessment of kidney function consists of using
the Glomerular Filtration Rate (GFR) which usually measured by creatinine clearance. But
serum creatinine is not a real-time marker of change in kidney function. It is neither a
specific nor a sensitive marker of GFR evaluation. In the case of rapid changes in kidney
function serum creatinine has a 48-72 hours delay related to GFR changes. It cannot show
the small changes of GFR too, and other factors rather than GFR may affect the blood level
of serum creatinine. Even in patients with stable serum creatinine the GFR may significantly
be declined. So many investigators are trying to redefine the cutoff point level of serum
creatinine increment. For instance the recent definition of AKI, considers a 0.3 mg/dl
increase of serum creatinine as an evidence of AKI development rather than a 0.5 mg/dl
increase (Shah & Mehta, 2006). Weisbord etal showed that only a 0.25-0.3 mg/dl increase in
serum creatinine after coronary angiography was accompanied with significant increase of
mortality within 30 days of procedure (Weisbord etal., 2006). In another study only a 5%
increases of serum creatinine within 12 hours after the contrast exposure had 75% sensitivity
and 72% specificity for detecting CIN. It also had a strong correlation with the development
of renal failure during next 30 days (Ribichini etal., 2010).
Recently more sensitive markers of GFR have been developed and validated. The most
promising markers are Cystatin-C and Neutrophilic Gelatinase Associated Lipocaline
(NGAL). Cystatin-C may be more accurate than serum creatinine in predicting renal
function. In patients with GFR lower than 60ml/min, Cystatin-C may be a better marker of
both early detection of CIN and prediction of major side effects like death and chronic
124 Advances in the Diagnosis of Coronary Atherosclerosis

dialysis. But it is not known whether the increased level of Cystatin-C is due to the
decreased GFR or it is released from atherosclerotic plaques during angiography .If
Cystatin-C increases by more than 10% within 24 hours, then it is a good marker of CIN
occurrence and further events. But an increase of less than 10% from baseline will rule out
the diagnosis of CIN and the patient can be discharged without any caution (Briguori etal.,
2010).There is no study to evaluate the validity of Cystatin-C in earlier time. Cystatin-C has
to be better validated in different situations.
NGAL is also an early diagnostic biomarker of acute kidney injury, but studies for
validating this biomarker in CIN are limited. It’s specificity and sensitivity in diagnosis of
CIN in children has been identified (Hirsch etal., 2007).
The most accurate method for measuring GFR is radionuclide techniques which are
expensive and labor-intensive. The Cockcroft-Gault or MDRD equations are useful for the
calculation of GFR in clinic.
The problem with Studies, regarding CIN is that nearly in all these studies contrast
nephropathy is defined by serum creatinine increment which is a soft outcomes value. If
hard outcomes like the need for dialysis, readmission, other clinical complications and death
are considered, it may cause better understanding the nature of AKI including CIN and
therefore its better management. It may also be more useful if RIFLE classification is taken
into account rather than Acute Kidney Injury Network criteria for better definition of CIN.
Further studies should be done to validate this classification for CIN diagnosis.
The differential diagnosis of CIN is cholesterol atheroemboli, volume depletion and
interstitial nephritis.

2.3 Incidence
CIN is the third cause of AKI in hospitalized patients. The cumulative incidence of CIN is
10% of all AKI (Tepel etal., 2006). The incidence of CIN in patients without any risk factor is
0.6-2.3%, but in high risk patients it will be raised up to 90% (McCullough etal., 2006A ;
Toprak & Cirit 2002). In two studies in 1979 and 2002, CIN was the third cause of in-hospital
AKI (Nash etal., 2002). These studies showed no change in the incidence of CIN in this
period (12% in 1979 and 11% in 2002).
Three factors increase the incidence of CIN in future: 1) increase in burden of CKD, 2)
Increase in incidence of diabetes, and 3) increase in diagnostic and therapeutically measures
requiring CM.
The incidence of CIN is dependent on the renal function. With serum creatinine more than
1.3mg/dl and more than 1 mg/dl in men and women respectively the risk of CIN will
significantly increase (Lamiere etal., 2006). It has been shown that by increasing serum
creatinine from 1.2 to 2.9mg/dl the risk of CIN increases from 4% to 20% (Barrett etal.,
1992;). The incidence of CIN in patients with serum creatinine of 3 mg/dl with or without
diabetes is more than 30%.In another study the risk of CIN in patients with serum creatinine
equal to1.5mg/dl was 8%, but with serum creatinine of 6.8mg/dl the risk of CIN has been
increased to 92% (McCullough etal.,1997). In patients with normal renal function, even in
the presence of diabetes or other risk factors the incidence of CIN is less than 2% ( Lamiere
etal., 2006). The incidence of CIN in diabetic patients with normal serum creatinine is 3.7%
(Rihal etal., 2002).In patient with severe renal failure and diabetes the need to dialysis will
be increase to 12% if CIN develops after CM exposure (Manske etal.,1990).
With regards to coronary angiography the incidence of CIN after this procedure has been
reported to be 3-14%. Only 2% of patients without diabetes and with serum creatinine of 1.1
mg/dl will develop CIN after coronary angiography (McCullough etal.,1997; Rihal etal., 2002).
125
Risks and Complication of Coronary Angiography: Contrast Related Complications

2.4 Natural course


The natural history of CIN includes an increase of serum creatinine mostly after 24 hours of
CM exposure, which picks in 3-7 days and return to normal within 2 weeks. Most patients
are non-oliguric but the urine output is usually declined (Rudnick etal., 1995).
CIN is generally and potentially a reversible acute renal failure, but it should not be
considered as a benign disorder, because in 30% of patients, renal function would not be
fully recovered (Maydoon etal., 2001). The patients with CIN rarely need dialysis. According
to a study among 0.8% of patients who needed dialysis after PCI, 13% remained on dialysis
for lifetime (McCullough etal., 1997). Another study reported that 13-50% of CIN patients
who had needed renal replacement therapy, they may remain on dialysis for the rest of life
(Toprak, 2007). If even CIN recovered completely, it will cause more morbidities including
bleeding, infection, prolonged hospital stay, increased resource utilization, increase in the
risk of CKD and increased in-hospital and long term mortality rate (Gruberg etal., 2000;
Perazella, 2009; Rihal etal., 2002). CIN also causes increase in cardiovascular morbidities.
The rate of both in-hospital and long term mortality will also significantly be increased due
to CIN (Gruberg etal., 2000; Liss etal., 2006; Marenzi etal., 2004; McCullough etal., 1997;
Rihal etal., 2002, Weisbord etal., 2006). The overall mortality rate of patients with CIN is 1.1-
34%, but it is not obvious that CIN perse is the cause of mortality or CIN is more prevalent
in patients who are sicker and their co-morbidities are more than control patients
(McCullough etal., 1997 Rudnick & Feldman, 2008).
The in-hospital mortality rate of CIN is estimated to be 22-25%. Meanwhile the rate of death
within 5 years is significantly higher in this group of patients in comparison with control
group (McCullough etal., 1997; Rihal etal., 2002). Odds of in-hospital death are related to
rate of serum creatinine increment (Weisbord etal., 2006). CIN is a cause of CKD in the
coming months (Liss etal., 2006). Although it is not known that whether the direct
nephrotoxic effect of contrast agents is the cause of CKD or the background renal disease
will be unmasked by CIN.

2.5 Risk factors


The risk factors of CIN are shown in table 2. Previous chronic kidney disease (CKD) is the
leading and the single most important risk factor of CIN (McCullough etal., 2006B; Rihal
etal., 2002; Rudnick etal., 1995). The most important cause of delaying or cancelling
angiography is renal failure. In a recent cohort, the Glomerular Filtration Rate (GFR) of 60
ml/min considered as a cutoff point (Bartholomew etal., 2004; Mehran etal., 2004). Diabetes
is the second most important risk factor of CIN (McCullough etal., 1997,2006; Rudnick etal.,
2006; Weisberg etal., 1994). It remains unclear whether diabetes is a risk factor of CIN perse
or it makes the patient prone to this complication because nearly all diabetic patient have
overt or covert renal disease (Manske etal., 1990). So the diabetic patients with CKD are the
highest risk group (McCullough etal., 2006aTepel etal., 2006; Toprak & Cirit, 2006).
Class IV heart failure is another risk factor of CIN (McCullough etal., 2006B). Many of these
patients also have several other risk factors like renal disease (atherosclerotic renal disease)
or decreased effective circulating volume and low GFR. Acute myocardial infarction (AMI)
is another independent risk factor of CIN if the CM is given within 24hours after the AMI
(Rihal etal., 2002; Marenzi etal., 2006a). S-T elevation myocardial infarction (STEMI) also
increases the risk of CIN in the time of PCI. The mortality rate of these patients is high too.
There is a positive correlation between the mortality rate of these patients and the volume of
126 Advances in the Diagnosis of Coronary Atherosclerosis

contrast that is being used for coronary angiography (Marenzi etal., 2006b). Old age
(Iokavou etal., 2003), hypovolemia (McCullough etal., 2006B) and hypotension are other risk
factors of CIN. Hypovolemia is among few risk factors which are preventable and should be
corrected promptly before starting the procedure. The medications that cause extra-cellular
contraction like diuretics or cause vascular tonicity changes like non-steroidal anti-
inflammatory drugs (NSAIDS) may be involved in development of CIN or at least may
increase the severity of the disease. The use of angiotensin converting enzyme inhibitors
(ACIs) or angiotensin receptor blockers (ARBs), while the blood pressure is in normal range
are not considered contraindicated and could be continued.

Patient related factors Procedure related factors


CKD hyper-osmolal CM
Diabetes mellitus high volume of CM
Age repeated exposure to CM within 24 hours
Hypovolemia intra-arterial injection of CM
Hypotension
Low ejection fraction
Class IV CHF
Concomitant use of other nephrotoxics
Hypoalbumenimeia (<3.5g/dL)
Renal transplantation
Recent AMI (24 hours before angiography)
Anemia
_________________________________________________________________________________
Table 2. Risk factors for CIN
Low hematocrite is associated with increased risk of CIN (Nikolsky etal., 2005), but there is
no study to show that correction of anemia will decrease the incidence of CIN.
There are few retrospective studies showing high incidence of CIN in renal transplanted
patients (up to 21%), but these patients had other risk factors like diabetes, renal failure,
concomitant use of nephrotoxic agents and volume depletion (Ahuja etal., 2000). The
coronary artery bypass graft (CABG) at the day of angiography will also increase the risk of
CIN (Ranucci etal., 2008). Nowadays in contrast to what is mentioned in the literature
multiple myeloma is not considered as a risk factor for CIN.

2.6 Type of CM
The safety of a contrast agent depends in large part on the amount of CM that must be
administered. The body’s ability to tolerate any substance depends on the amount given.
Even isotonic saline is toxic at very large doses. High dosages that disturb the ionic and
osmotic balance in the body will elicit adverse reactions without exhibiting a direct
interaction with other molecules (chemotoxicity). This kind of toxicity is based on the
osmotic activity of the agent or, more precisely, on the osmotic load (mosmol/kg body
weight). The important properties of a CM that determine its nephrotoxicity are: osmolality,
volume, and repeated use, route of use, viscosity and Ionicity.
Here we briefly review the role of each of the properties of a CM in the development of
CIN.
127
Risks and Complication of Coronary Angiography: Contrast Related Complications

2.7 Osmolality / viscosity (Table2)


Now the HOCM is completely replaced by LOCM or IOCM. There is no doubt that
osmolality is a main factor in nephrotoxicity of a CM (Barrett & Carlisle, 1993; Rudnick &
Goldfarb, 2003). Although in patients with normal renal function this factor is not important
(Barrett & Carlisle, 1993). But in chronic renal failure patients, particularly in diabetic
patients, use of HOCM is contraindicated (Solomon, 2005, Solomon & Dumouchel,
2006).The incidence of CIN with HOCM in patients with risk factors is two times in
comparison with LOCM (Barrett & Carlisle, 1993).
All studies showed that nephrotoxicity of low osmolal media is lower than hyperosmolal
one (Barrett & Carlisle, 1993; Rudnick etal., 1995), but there are many controversies about
the difference of LOCM and IOCM in terms of CIN (Aspelin etal., 2003; Barrett etal., 2006;
Carraro etal., 1998; Liss etal., 2006; McCullough etal., 2006C; Rudnick etal.,2006; Solomon
etal.,2007; Thomsen etal., 2008).The viscosity of IOCM is higher than LOCM and this
property of IOCM may offset the benefit of lower osmolality of newer developed agents
(Aspelin etal., 2003; Barrett & Carlisle, 1993; Rudnick & Goldfarb, 2003, Seeliger etal., 2007).
Systematic reviews were not also able to solve the contradictory results concerning the
difference between low and iso-osmolal CM (Heinrich etal., 2009, Reed etal.,
2009,McCullough etal., 2006C., Solomon & Dumouchel, 2006). NEPHRIC study showed that
with IOCM the risk of CIN in high risk diabetic patients is significantly lower than LOCM
(Aspelin etal., 2003). This study is criticized by few numbers of patients. In a meta-analysis it
was also shown that IOCM are less nephrotoxic than LOCM (McCullough etal,. 2006C), but
in another larger meta-analysis there was no significant difference between LOCM and
ICOM in term of nephrotoxicity (Heinrich etal., 2009). In a retrospective study, LOCM was
less nephrotoxic than IOCM (Liss etal., 2006). This study showed that the risk of renal failure
in future is increased by IOCM whether they had CIN or not.
According to the published studies, the American college of cardiology/American Heart
association (ACC/AHA) recommended that there is no difference between low and iso-
osmolal agents in CKD patients. The exception is Iohexol and Ioxaglate. The IOCM has
lower probability of CIN than these two LOCM (Kushner etal., 2009).
The exact role of Ionicity in development of CIN is not known. The only meta-analysis in
this regard showed that Ionic IOCM is similar to nonionic LOCM (Barrett & Carlisle,
1993).

2.8 Contrast Media volume


There is a direct relation between dose of CM and CIN development (Manske etal., 1990;
McCullough etal., 1997; Rudnick et al., 1995). The probability of CIN is increased with
higher volume of CM. On average, in coronary angiography, PCI and CT scan 130cc, 190cc
and 100 to150cc CM is used. Although it has been shown that no patient with less than 100cc
CM has developed CIN (Gruberg etal., 2001; McCullough etal., 1997), but unfortunately
wide range of the volume of CM defined as cut off value (30-200cc). The relation between
the dose of CM and CIN is particularly evident in moderate to severe renal failure patients
(CKD3-5) which they should be received ultra-low dose of CM. Unfortunately, the need for
CM for interventional purposes is much higher than this safe volume (250-300cc). In the
study of Rihal etal it has been shown that with each 100cc CM, the risk of CIN will be
increased 12% (Rihal etal., 2002). In another study the risk of CIN is serious when the
volume of CM exceeds more than 3.7 fold of creatinine clearance (Laskey etal.,
2007).Repeated use of CM within 72 hours increases the risk of CIN
128 Advances in the Diagnosis of Coronary Atherosclerosis

3. Pathogenesis
The primary mechanism of CIN is Ischemia and direct tubular toxicity of CM to epithelial
tubular cells.In experimental studies, after contrast infusion there is an early brief
vasodilatation of renal vasculature, followed by a long term vasoconstriction (Arakawa etal.,
1996; Seeliger etal., 2007). This biphasic response has not been evident in human and mostly
increase of renal blood flow has been shown (Weisberg etal.,1992). These studies carried out
the direct measurement of blood flow by hemodilution techniques. Nevertheless, if renal
blood flow is measured directly by an intra-renal artery guide-wire, no difference of blood
flow could be detected at the time of ventriculography, while subsequently by further CM
infusion, the blood flow is decreased by3.7-39.5% from base (Mockel etal., 2008).Although
these studies showed mild to moderate decrease of renal blood flow after exposure to CM,
these findings nevertheless cannot describe the longitudinal decrement of GFR in CIN
patients.

3.1 Medullary Hypoxemia


To overcome the discrepancy between renal blood flow rate and GFR decrement, medullary
Hypoxemia due to regional blood flow changes is in the focus of attention in describing the
pathophysiology of CIN. According to this hypothesis, the pathogenetic axel of CIN is
medullary hypoxemia (Agmon etal., 1994; Heyman etal, 2008).
Physiologically, blood flow in outer medulla is low and this part of nephron works in
relatively hypoxic state. The cause of this condition is the unique structure of vasa recta
which is vital for countercurrent solute exchange. Low blood flow of medulla is also
important to maintain the gradient in distal nephron. Meanwhile the thick ascending Limb
of Henle as a hard worker of nephron needs a high level of oxygen. Due to this
contradictory status, this part of nephron is more vulnerable to ischemic injuries. The
decrease of blood flow in outer medulla is critical. In normal physiological conditions, the
mechanisms responsible for blood flow and tubular transport are tubule-glomerular
feedback, autoregulation and glomerulo-tubular balance which operate hand in hand to
maintain the integrity of nephron and the system to work properly. The vasodilators (nitric
oxide, prostaglandins, adenosin, dopamin, urodilantin) and vasoconstrictors (local
endothelin, AngiotensinII, vasopressin) and transport inhibitors (PDF2, Adenosin,
dopamine) together balance the supply and demand of oxygen in this part of nephron.
Release of vasoconstrictor mediators or blockade of vasodilator mediators may play a role in
the pathogenesis of CIN (Heyman etal., 1994). CM makes imbalance of these mediators.
They cause decrease of oxygen tension both in cortex and medulla. It may be due to
increased work of active transport in medullary tick ascending limb of Henle in response to
osmotic diuresis of hyperosmolal agents.
Proximal tubules reabsorb a scant amount of CM. The osmotic property of CM causes less
water reabsorption and increased intra-tubular pressure. The distal delivery of salt and
water will be increased in this state which in turn activates tubule glomerular feedback and
decreased GFR.
Increased pressure of intrestitium is also a factor for decreasing GFR. It also causes
medullary hypoxia. CM may decrease renal blood flow by direct vasoconstriction of renal
vasculature. This effect may be due to the change of calcium metabolism (Bakris & Burnett,
1985), or local increase of adenosin (Pflueger etal., 2000) and endothelin (Bagnis etal., 1997).
By CM infusion, the oxygen tension in outer medulla is decreased, nevertheless the sodium
delivery to thick ascending Limb of Henle is increased due to osmotic diuresis of CM but
129
Risks and Complication of Coronary Angiography: Contrast Related Complications

the low oxygen pressure in this part of nephron is not related to the osmolality of CM (Liss
etal., 1998). Administration of A1 adenosin receptor antagonists could not humiliate the
decreased oxygen tension (Liss etal., 2004), but it seems that prostaglandins and nitric oxide
could counteract vasoconstrictor effect of CM (Agmon etal., 1994). In animals, nitric oxide
depletion has an effective role in vasoconstriction (Agmon etal., 1994; Ribeiro etal., 2004).

3.2 Reactive oxygen species


The role of oxidative stress in pathogenesis of Ischemia/reperfusion injuries is well known.
Activation of oxidative stress processes in ischemia produce reactive oxygen species (ROS)
which have a known role in AKI. The diseases which are considered as risk factors of CIN
(CKD, CHF, and Diabetes) are associated with high oxidative stress state.
Experimental studies demonstrated that therapeutical strategies reduce the production of
ROS and/or increase the nitric oxide activity and by these mechanisms halt the renal
injuries due to Ischemia in different models of AKI including CIN. The rational use of
antioxidants such as acetylcystein or allopurinol in preventing CIN is to lower or inhibit the
oxidative stress processes.

3.3 Direct cytotoxicity


It is not known how much physicochemical properties of Iodinated CM are responsible for
CIN and whether direct cytotoxicity of CM play any role in pathogenesis of CIN or not. In
vitro studies showed that CM particularly HOCM is cytotoxic to mesangial and tubular cells
(Itoh etal., 2006; Peer etal., 2003). The mechanism of such cytotoxicity is not known, but
possibly cellular energy failure, disintegration of calcium homeostasis, apoptosis
mechanisms and oxidative stress play a role (Beeri etal., 1995; Haller & Hizoh, 2004; Humes
etal., 1987;). Internalization of non-ionic agents are seen in proximal tubules, (nephrosis
osmotic), but these pathologic findings are also seen in patients who take CM without renal
function deterioration (Persson & Tepel, 2006).CM may cause vasoconstriction by direct
action on smooth muscles of arteries. More studies, should be run to show the relation of
these changes with iso-osmolal contrast agents.

3.4 Others
In experimental studies CM can decrease capillary blood flow by decreasing the velocity of
erythrocytes and increasing the red blood cells aggregation. HOCM also decreases
erythrocyte volume and alters its membrane deformability. Recently hemeoxygenase
activator is introduced as a pathomechanism of CIN development. Hemeoxygenase inhibits
ROS production, suppresses pro-apoptotic proteins, activate anti-apoptotic proteins and has
anti-inflammatory and vasodilatory effects. In future this enzyme may play an important
role in CIN prophylaxis in human.

4. Prophylaxis
If CIN happens, the management is supportive, and because the time of insult is known the
best measure is prophylaxis. Although in many studies, meta-analysis and systematic
reviews have focused on prophylactic measures of CIN, nevertheless there is no an
evidence-based guideline for clinical use.
130 Advances in the Diagnosis of Coronary Atherosclerosis

No general consensus is present about the prophylactic measures of CIN other than
stopping nephrotoxic agents, hydration and use of the lowest possible volume of CM and
not using HOCM. Published guidelines are according to limited studies on very heterogenic
patients. So they cause more confusion rather than better understanding and enlightment of
the issue of CIN to provide the best reliable prophylactic measures. Probably the European
Society of Urogenital Radiology (ESUR) guideline is yet valid (Morcos etal.,1999) and
further studies within last 13 years have not been demoted its validity.

4.1 Volume expansion


Theoretically, volume expansion suppresses renin-angiotensin system activity, decreases
vasoconstrictor hormones like endothelin, increases sodium diuresis, inhibits tubulo-
glomerular feedback, prevents tubular obstruction, protects cells against ROS and finally
dilutes the CM in tubules. Overall the toxicity of CM is decreased on tubules by hydration.
Large numbers of studies are about the role of volume expansion in preventing CIN. No
controversy is present about the effectiveness of volume expansion. Up to now the volume
expansion is known as the most effective measure in nephroprotection of CIN (Thomsen,
2006).This benefit has been shown in many randomized controlled trials (Bader etal.,2004;
Dussol etal., 2006; Merten etal., 2004; Mueller etal., 2002; Taylor etal, 1998; Trivedi etal.,
2003). But it is difficult to show that which type of fluid in how much dose and at which
time is more suitable (McCullough & Soman, 2005). This obscurant is due to the
heterogeneity of the studies and different definitions for CIN are used. Unfortunately these
studies rarely reported the hard clinical endpoints.
Solomon et al has done the first randomized controlled trials (RCT) in this regard (Solomon
etal., 1994). They showed that forced diuresis with diuretic or manitol is worse than
hydration with isotonic salin or 0.45% salin. This Study did also very clearly show that
volume expansion is highly effective in prevention of CIN in high risk patients and since
then this prophylactic measure is the key stone of CIN prevention. Intravenous fluid
therapy is also superior to oral hydration (Weisbord & Pavelsky, 2008) as it was shown in
the study of Solomon etal .They reported that the control group had had 10 times more CIN
than case controls. Because of such significant difference, this study was stopped early.
Mueller etal also showed superiority of isotonic saline to 0.45% salin in patients with normal
renal function, but if renal failure is present, there is no difference between two types of
fluids (Mueller etal., 2002). The study of Merten etal compared sodium bicarbonate with
isotonic saline in preventing CIN. Although this study showed better results with
bicarbonate, but the study is criticized in different aspects (Merten etal., 2004).
In some studies intravenous bolus of isotonic saline just before procedure, or oral hydration
from the night before the procedure, or use of NaCl tablets from 2 days before contrast
exposure were examined with different results. (Bader etal.,2004; Dussol etal., 2006; Taylor
etal, 1998; Trivedi etal., 2003) Volume expansion with isotonic saline is safe, simple and
cheap, but not all patients taking CM will unfortunately suffer it. We showed that in
patients that use of isotonic salin may be associated with the hazard of overload, use of
0.45% salin may be as effective as isotonic salin (Vasheghani-Farahani etal., 2010). It has
been demonstrated that 26% of high risk patients have not been received any kind of
intravenous fluid, and there was no standard about the volume and the type of fluid that
were being used in the remaining patients. Urine volume more than 150cc / hour is shown to
be a good sign of decreased risk of CIN (Stevens etal., 1999). Cardiologists are the main
target group who should know and use the strategies of volume expansion.
131
Risks and Complication of Coronary Angiography: Contrast Related Complications

4.2 Sodium bicarbonate


The mechanism of sodium bicarbonate in preventing CIN is not clearly understood.
Theoretically Sodium bicarbonate can expand the extracellular volume.
Alternatively it has been suggested that sodium bicarbonate will decreased the amount of
ROS by increasing the PH of medulla. So the kidney will be protected from oxidant injuries
(Atkins, 1986; Bakris etal., 1990; Merten etal., 2004; Morcos etal., 2005; Weisbord etal., 2008).
Through this line of explanation, the main protective effect of sodium bicarbonate is not
through volume expansion.
There are many concerns about the role of bicarbonate in prevention of CIN (Briguori etal.,
2007; From etal., 2008; Masuda etal., 2007; Ozcan etal., 2007, Recio-Mayoral etal., 2007).Most
studies compared isotonic saline and sodium bicarbonate have reported contradictory
results. Masuda etal., showed Bicarbonate is more effective than saline in prevention of CIN
(1.7% Vs 13%) (Masuda etal.,2007). In a meta-analysis, comparing saline and bicarbonate, it
was shown that rise of serum creatinine in bicarbonate group is less than salin group, but in
term of need to dialysis and death there was no difference between the two groups(Hogan
etal., 2008).In the largest RCT comparing salin and bicarbonate, No differences between two
groups was shown (Maioli etal., 2008). In another retrospective study including 7977
patients it was shown that the risk of CIN in sodium bicarbonate is significantly higher than
salin group (From etal., 2008).We showed that addition of sodium bicarbonate to isotonic
salin added no benefit to isotonic salin alone in preventing CIN (Vasheghani-Farahani etal.,
2009).These controversial results and heterogeneity of studies make it difficult to be able to
extract a conclusion that bicarbonate is even equal to salin in preventing CIN.

4.3 N-Acetyl Cysteine (NAC)


The main mechanism of NAC may be due to its known antioxidant peculiarity. It may
diminish the oxidative stress markers ((Drager etal., 2004; Efrati etal., 2003) and / or its
vasodilatory effect (Fishbane etal., 2004; Stenstorm etal., 2008).CM decrease urinary nitric
oxide and NAC may inhibit this effect of CM. But it has been shown that NAC has no effect
on F2- isoprostane a known marker of oxidative stress (Efrati etal., 2003).
NAC has wide first-pass metabolism, so its bioavailability will be very low if it is
administered orally. Meanwhile many centers use oral NAC without any evidence-based
guidelines (Marenzi etal., 2006b). The drug has also wide intra-patient variation. Its half-life
after intravenous injection is 40 minutes. It has wide plasma and tissue protein binding.
After oral or intravenous administration of NAC the drug cannot be detected in systemic
circulation. So its effectiveness may be due to secondary effects like induction of glutathione
synthesis rather than its direct effect. Because there are no tool to measure these secondary
effects, no optimal dose of NAC can be defined and the dose of drug for prevention of CIN
is empirical.
Clinically the use of NAC was controversial from the early time. Positive and negative
results about NAC have created confusions about the role of this agent in CIN prophylaxis
protocols. When the first report of NAC was published by Tepel and his coworkers (Tepel
etal., 2000), it caused a universal trend to use NAC in all patients have been taking CM.
They showed significant decrease in the rate of CIN in patients on NAC (2% Vs 12%). Since
then many studies have been conducted with contradictory results. Now After 12 years of
use of NAC in prevention of CIN, there is no general consensus about this pharmacological
agent. In last decade many RCTs, non-randomized studies and meta-analyses were
performed with NAC (Alonso etal., 2004; Bagshaw etal., 2006, Bagshaw & Ghali, 2004; Birck
132 Advances in the Diagnosis of Coronary Atherosclerosis

etal., 2003; Duong etal., 2005; Dzgoeva etal., 1995, Isenbarger etal., 2003; Kshirsagar etal.,
2004; Liu etal., 2005; Nallamothu etal., 2004; Onbasili etal., 2007; Pannu etal., 2004; Spargias
et al., 2004; Zagler etal., 2006). Some of these studies showed that there is no positive effect
for NAC in prevention of CIN (Onbasili etal., 2007; Spargias et al., 2004). On the other hand
some meta-analyses showed effectiveness of NAC but the most of these meta- analyses had
inconclusive results (Bagshaw etal., 2006; Nallamothu, 2004). In conclusion although huge
data is available, the role of NAC in preventing CIN is moot yet. This discrepancy may be
due to bias in the publishing of studies with positive results, so these studies should be
considered with skepticism (Fishbane etal., 2008).
In a study on patients with myocardial infarction high dose intravenous NAC (1200mg) was
effective in prevention of CIN (Marenzi etal., 2006b). Surprisingly no patient in this study
had received fluid for volume expansion. An interesting but confusing issue in studies about
NAC is the decrement of serum creatinine after CM exposure (Baker etal., 2003; Drager etal.,
2004; Kay etal., 2003; Marenzi etal., 2006b). Probably NAC can decrease serum creatinine
directly without affecting renal function (Drager etal., 2004). In this study the GFR of
patients who were taken NAC four days before CM exposure was significantly higher than
those patients were taken NAC only for two days. Interestingly this study and study of
Hoffmann and coworkers showed that NAC may decrease serum creatinine in patients that
had not received CM (Hoffmann etal., 2004). There are concerns that NAC with increasing
tubular secretion of creatinine decreases the serum creatinine without any change in GFR. A
recent meta-analysis found that NAC was more protective than saline infusion and the
authors recommended the use of NAC because of its low cost, availability and paucity of
side effects (Cigarroa etal., 1989). In contrast to this recommendation which is based on
safety of the NAC, there is a report of anaphylactic reaction in 3-6% of patients receiving
intra-venous NAC (Kanter, 2006).
Briefly, NAC may increase GFR in normal patients and restore GFR in high risk patients for
CIN. But it is not known whether these effects are due to direct protective effect of NAC on
kidney or it is due to alteration in creatinine metabolism. Some studies reported good effect
of NAC while others did not find such positive effects.
In conclusion, there is no agreement about the NAC. Otherwise there is no guideline about
the route of administration of NAC or the dose or the time of its administration. There is an
important question whether its effect on serum creatinine is real or spurious. It is obvious
that we need more good designed RCTs to solve these controversies.

4.4 Type and dose of Contrast Media


It is obvious that HOCM is associated with higher risk of CIN. So for prophylactic purposes
it is recommended not to use this type of CM in any patients particularly in the patients
with moderate or high risk of CIN. In analysis of present studies about the type of contrast,
it is possible that different monomer CM have different toxic effects (Bettmann, 2005;
Solomon etal., 2005). Up to now there is no evidence that CIN is more prevalent with LOCM
than IOCM. There is a trend to use Iodixanol which is a dimeric IOCM (Barrett & Carlisle,
1993). But different studies showed no superiority of this agent over the other IOCM or
LOCM (Baker etal., 2003; Boccolandro etal., 2003; Briguori etal., 2005). Over all, it seems that
there is no difference between LOCM and IOCM in regard of CIN.
The other issue in preventing measures for CIN in the term of CM is the dose of the agent.
So it is advised that the lowest possible dose of the CM should be administered in patients at
risk of CIN. It is obvious that the higher the volume of the CM is associated with the higher
133
Risks and Complication of Coronary Angiography: Contrast Related Complications

incidence of CIN (Cigarroa etal., 1989; Manske etal., 1990), but there is no magical dose of
contrast to believe that below that dose, the CIN will not occur. The renal function is an
important limiting factor for dose of CM. In study of Laskey etal. it was reported that if the
volume of CM exceeded 3.7 times of creatinine clearance, the risk of CIN will be higher
(Laskey eal., 2002). So the absolute value of contrast volume may be too low. There are 2
formulas for calculating relatively the safe volume of CM according to renal function.

ml
5     contrast  media
kg
dose of CM    mg (1)
serumcreatinine  ( )
dl
Or,

300 ml  contrast  media    


 (2)
serumcreatinine
 
In any case the dose of contrast should not be exceeded by 300 ml. For IOCM and LOCM the
dose may increase by 1.5 times.

4.5 Other preventive strategies


Several studies examined different pharmacologic agents to prevent CIN. Studies about
fenoldopam, dopamin, calcium channel blockers, atrial natriuretic peptide, L-arginine and
statines are inconclusive. Use of ATP-MgCl2 also failed to show beneficial effect. Mesna is
another pharmacologic agent that can inhibit ROS related renal injury.

4.5.1 Theophylin
High level of adenosin is a possible mechanism for occurrence of CIN. It causes afferent
arteriole vasoconstriction (Haller etal., 2004; Pflueger etal., 2000). Theophylin is a non
specific antagonist of adenosin receptor, so it may theoretically be beneficial. Studies about
the theophylin like studies of NAC are not conclusive. Few studies showed effectiveness
of theophylin or aminophylin in prevention of CIN , but in a meta-analysis no positive effect
of this drug was shown (Bagshaw & Ghali, 2005). So the routine use of theophylin is not
recommended. In a study on patients in ICU, infusion of 200mg theophylin half an hour
before contrast exposure was superior to NAC (Huber etal., 2006) but arythmogenesity of
theophylin is a problem in coronary artery disease. It is possible that in the future,
theophylin is replaced by a selective adenosin receptor antagonist with less toxicity and
more effectiveness than theophylin in CIN prophylaxis protocols (Pflueger etal., 2000).

4.5.2 Prostaglandin analogues


Prostaglandin I1 and E1 may have some protective effects , but it is associated with severe
hypotension. So their routine use is not suggested. Prostacyclin may also have such a role.

4.5.3 Ascorbic acid


Studies about the ascorbic acid are inconclusive (Briguori etal., Spargias etal., 2004). 3 grams
of ascorbic acid 2 hours before contrast exposure and 2 grams at night and in the morning
after procedure may decrease the risk of CIN (Spargias etal., 2004).
134 Advances in the Diagnosis of Coronary Atherosclerosis

4.5.4 Trimetazidine
This novel pharmacologic agent interferes with different metabolic pathways. It can prevent
Ischemia-reperfusion injuries. In a RCT it was effective in preventing CIN (Onbasili etal.,
2007).

4.5.5 Erythropoietin
Erythropoietin may have some role in CIN prophylaxis. Its effect has been shown in some
studies (Yokomaku etal., 2008).

4.6 Renal replacement therapy


The rationality for use of renal replacement therapy after contrast exposure is to remove the
CM rapidly to prevent CIN (Frank etal., 2003; Marenzi etal 2003,2006b; Vogt, 2001).Each
session of dialysis can remove 60-90% of administered CM (Deray etal., 2006). CM are
totally eliminated by the kidneys. They are handled by the kidneys like Inulin. Different
extracorporeal techniques can efficiently remove CM. So it is rational to use these techniques
just after contrast exposure to prevent or reduce the risk of CIN. Recent study on severe
CKD patients not on dialysis(GFR less than 15ml/min) comparing isotonic salin and
prophylactic hemodialysis showed that patients on hemodialysis were discharged from the
hospital while 5% of them need hemodialysis. Meanwhile, the rate of chronic dialysis at the
time of discharge in salin group was 45% (Solomon etal., 2004). Nevertheless, other studies
demonstrated that hemodialysis in patients with previous history of renal failure was not
beneficial and the incidence of CIN will not be changed by this modality (Deray, 2006; Vogt,
2001).One meta-analysis has supported this result (Cruz etal., 2006). Patients on dialysis do
not also need dialysis after contrast exposure (Hamani etal., 1998; Morcos etal., 2002).
Only few study reported the prophylactic usefulness of hemodialysis (Lee etal., 2007;
McCullough & Soman, 2005). But repeatedly it has been shown that hemodialysis has no
role in preventing CIN (Cruz etal., 2006; deray, 2006; Marenzi etal., 2006b). In contrast to
positive studies one RCT showed that prophylactic dialysis caused more reduction of renal
function and these patients need more dialysis after exposure to contrast (Vogt etal., 2001).
In one meta-analysis (Cruz etal., 2008), the beneficial effect of pre-procedural extracorporeal
blood purification was obscure.
It has been shown that serum creatinine four days after contrast exposure is less in dialysis
group, but this difference was not significant.
CRRT, hemodiafiltration (CVVHD) and hemofiltration are also evaluated in this regard
(Gabutti etal., 2003, Marenzi etal., 2003). The only technique with enthusiasm was CVVH.
But this procedure is very complex and expensive and even if further studies prove its
efficacy in large clinical trials, it cannot be ordinarily used in clinic (Klarenbach etal., 2006).
Overall, given the cost and logistic necessity of extra corporeal modalities, it is difficult to
suggest routine use of these techniques in prophylactic protocols of CIN and further studies
may show its advantage or disadvantage.

4.7 Drugs which should be stopped before procedure


At the time of planning for procedures with CM, all drugs which are taken by patients
should carefully be reviewed. There is no RCT about the harmfulness of drugs, but
generally speaking the following drugs may be associated with increased risk of CIN.
135
Risks and Complication of Coronary Angiography: Contrast Related Complications

4.7.1 Non-Steroid Anti-Inflammatory Drugs (NSAIDs)


Case reports and clinical experiences have shown that NSAIDs may increase the incidence
of CIN. There is no difference between selective or non selective cycloxygenase inhibitors.
They should be stopped several days before procedure.

4.7.2 Antihypertensive agents


No antihypertensive drug is contraindicated for patients that undergo for imaging with CM.
But if they cause hypotension, the risk of CIN may be increased. The use of angiotensin
converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) should not
be stopped (Erley, 2006). Although In patients with serum creatinine 2 mg/dl or higher it
may increase the rate of CIN. Cirit etal. have reported an increased risk of CIN with ACEIs
(Cirit etal., 2006), nonetheless another study showed its preventive effect (Dangas etal.,
2005).There is also a report that Captopril may cause decrease in incidence of CIN in
diabetic patients(Gupta etal., 1999).
There is no study about ARBs.

4.7.3 Metformin
Metformin causes lactic acidosis in patients with renal failure. Otherwise uncontrolled
diabetes is a risk factor of CIN. There is no RCT about metformin and CIN. It is
recommended to continue the use of the drug until the night before the procedure (Bailey
etal., 1996). Others have suggested the continuation of metformin in patients with normal
renal function. If metformin is stopped, it is better to start it again when the physician is sure
that CIN has not been happened.

4.7.4 Diuretics
Use of furosemide, manitol and endothelial receptor antagonists were associated with
increased risk of CIN (Anto etal., 1981; Solomon etal., 1994; Weisberg etal., 1994).Forced
diuresis increases the risk of CIN. Although there is no RCT about diuretics, nevertheless it
is not theoretically rational to expand the volume from one hand and to contract it from the
other hand. So it is recommended to discontinue diuretics 12 hours before the procedure
and to restart it 24 hours after that.

4.7.5 Manitol
Like diuretics it may increase the risk of CIN in patients with forced diuresis (Solomon etal.,
1994). The problem is raised in neurosurgical patients on manitol. It is suggested to stop the
drugs several hours before CM administration and to monitor the hydration status of the
patients very carefully (Erley, 2006)

5. Tools for assessment of patients at risk of CIN


In published study there is no controversy about the risk factors of CIN. But nearly all
patients undergoing imaging with CM have more than one risk factor, so the investigators
are looking for an innovative risk stratification model to detect the patients who are at a
higher risk of CIN. There are different scoring tools that could detect the patients at risk of
CIN before the procedure and accordingly the physicians can provide and modify the
different preventive measures for these patients to decrease the risk of CIN.
136 Advances in the Diagnosis of Coronary Atherosclerosis

Up to the present time there are two popular scoring models. Both of which have been
introduced at 2004 and provide good tools for estimating the odds of the CIN. They are
more or less similar with few differences in the terms of risk factors and scoring. According
to the data of 8357 PCI patients, Mehran et al. determined the independent predictive factors
of CIN and provided a scoring model for estimating potential risk of patients (Mehran etal.,
2004). Bartholomew et al. also suggested another risk stratification model at the same year
(Bartholomew etal., 2004). They evaluated the data of 2047 patients and determined the risk
factors and their risk scoring. Table 3 shows these two models comparatively.
According to Mehran's model the risk score of less than 5 is associated with 7.2% risk of CIN
and nearly no patients need dialysis (0.04%) but risk score more than 16 will increase the
risk of CIN up to 57.7% and need to dialysis to 12.6%.
Bartholomew et al. reported no patients with risk score of less than 1 developed CIN and
CIN had occurred in 25% of patients with risk score more than 9.
In regards to risk stratification models, some factors are not predictable before the procedure
(like need to balloon pump or volume of contrast), so according to these models it is
impossible to estimate the exact reliable and accurate risk of CIN in every patient.

Mehran model Bartholomew model


Risk factor Point Risk factor point
Hypotension(BP<80 mmHg) 5 Hypotension 1
CHF 4 CHF 1
Intra-aorta balloon pump 5 Intra-aorta balloon pump 2
Diabetes 3 Diabetes 1
Volume of contrast 1(/ each100cc) Volume of contrast(>260cc/) 1
Previous CKD: Creatinine clearance<60ml/min 2
Serum Creatinine> 132 mmol/L 4
Or:
GFR 40-60 ml/min 2
GFR 20-40 ml/min 4
GFR <20 ml/min 6
Anemia 3 Peripheral vascular disease 1
Age > 75 years 4 Emergent PCI 2
Table 3. Two most popular risk starfication models of cin

6. Recommendations
Until more accurate and universally accepted guidelines are developed, following
recommendations which are based on different studies are useful in preventing CIN.The
important issue is how to find the patients at risk before imaging procedures. There are
some recommendations in this regard.
The American Radiology College (ARC) has suggested the measurement of serum creatinine
in all patients who are suspected of having renal disease or those at risk of nephrotoxicity
before the procedure (The committee on drugs and contrast media of the American College
of Radiology, 2010). The patients at risk are those with a positive answer to any of the
following statements: history of renal disease (including tumor or transplantation), familial
history of renal disease, on diabetes therapy, having paraproteinemia disorders, having
137
Risks and Complication of Coronary Angiography: Contrast Related Complications

collagen vascular disease, taking specific drugs such as metformin or NSAIDs or


Nephrotoxic antibiotics. The ESUR has also recommended to measure serum creatinine in
some specific patients (Thomsen & Morcos, 2005). Accordingly, the patients with a history
of renal disease, renal surgery, proteinuria, hypertension, diabetes, gout, recent use of
nephrotoxic drugs are considered high risk patients. The Urogenital Radiology Association
also suggests that serum creatinine should be measured in all patients with history of
chronic renal failure or those who are candidates for intra-arterial angiography or patients
with at least one positive answer to the questioner (Thomsen & Morcos, 2006). 99% of
patients with negative answers to all questions had serum creatinine less than 1.7 mg/dl
(Thomsen, 2005).A good alternative surrogate marker to the serum creatinine is to measure
GFR according to one of the MDRD formulas in all patients with any risk factors of CIN,
because the serum creatinine has limitation to find CKD patients.
After finding the high risk patients there are some different preventive measures for people
at different risks. But some general recommendations should be taken into account for all
patients whether they have risk factor(s) or not.

6.1 General recommendations for all patients taking CM


1. Evaluation of risks in all patients planned to take CM.
2. Measurement of serum creatinine in all high risk patients before and after
procedure.
3. List the risk factors according to present risk score models.
4. Encouraging all patients to drink water before procedure.
5. All patients should be taken LOCM or IOCM.
6. The dose of CM should be as low as possible.
7. Continuing use of angiotensin converting enzyme inhibitors and angiotensin receptor
blockers is not contraindicated
8. NSAIDS should be stopped several days before procedure.
9. Use of furosemide and manitol should be stopped 12-24 hours before CM
administration.
10. Stop metformin the night before procedure until ensure of not happening CIN.
11. No repetition of procedure until to be sure that CIN has not been developed.

6.2 Recommendations for low risk patients


(Normal renal function and risk score≤5 according to Mehran's score)
1. All recommendations for no risk patients
2. Oral hydration (at least 500cc pre-procedure and 2500cc within 24 hours post
procedure)
3. Intravenous isotonic salin in patients who are not taking oral intake at rate of 1
ml/kg/hour starting 4 hours before procedure and continuing at least for 6 hours post
procedure . It is best to continue the hydration for 24 hours after procedure.
4. No repetition of procedure until serum creatinine returns to base value.

6.3 Recommendation for moderate risk patients


(Patients with CKD1-3 and/or risk score 5-15 according to Mehran's score)
1. All recommendations for low risk patients
138 Advances in the Diagnosis of Coronary Atherosclerosis

2. Delaying the procedure in volume depleted patients or those who have blood
circulation collapse or patients with CHF until stability of hemodynamic is achieved.
3. Infusion of 0.45% salin in patients at risk of volume overload.
4. If the time to procedure is less than 6 hours (in emergent angiography) a simultaneous
administration of oral and IV Fluid with the dose of 3 ml/kg/hour and maximum 300cc
fluid/hour is recommended. The fluid administration should be started one hour
before procedure. After procedure the fluid is continued with the rate of 1 ml/kg/hr for
12 hours.
5. Use of NAC 1200mg twice daily in the day and day after the procedure. In emergency
situations use of 1200mg intravenous bolus of NAC instead of first oral dose is
appropriate.
6. Measurement of serum creatinine 24-48 hours after procedure in patients with
moderate and severe risk factors

6.4 Recommendations for high risk patients


(CKD4-5 and/or risk score >15 according to Mehran's score)
1. All recommendations in moderate risk patients
2. Nephrology consultation
3. Use of hydration should be individualized according to volume status of each patient
with considering overload hazards.
4. In ICU patients use of 200mg theophylin 30minutes before procedure, particularly if
hydration protocol could not completely be follow. But remember the side effects of this
drug.

7. Anaphylactic reactions
Anaphylactic reactions to CM are well known from the early use of these agents. These
reactions are dermal allergic type reactions, edema, pulmonary edema, angina pectoris,
arrhythmia and hypertension crisis. Mostly these side effects are mild and there is no need
to treatment. The occurrence of the serious side-effects is within 20 minutes of CM
administration. They are exhibited by nausea and vomiting, urticaria, pulmonary edema,
bronchospasm, laryngospasm, hypotension, tachycardia, vaso-vagal responses and seizure.
In last decade the reactions to CM have significantly decreased from 5-12% to 0.2-0.7%
(Sayol & Bush, 2011). It may be due to shift from HOCM to LOCM and IOCM. Anaphylactic
reaction to HOCM is more than LOCM. Ionicity is also a cause of reaction to these agents
and nonionic monomers are less anaphylactic. These reactions are either chemotoxic or
Anaphylactic (Idio or nonidiosynchratic).The chemotoxic reaction is related to
physicochemical property of CM and its severity is dependent on the dose and
concentration of the agent.
The mechanisms of anaphylactic reaction are not well understood. Histamine, Leukotriens,
Basophiles and prostaglandins may be mediators of this type reaction. It is unlike that
antigen- antibody interaction plays any role.
Aging, history of allergy and asthma and previous history of reaction to CM, are the risk
factors of anaphylactic reactions. The patients with CKD, cardiovascular disease and
epilepsy are also at increased risk. Beta blockers may increase the threshold and severity of
anaphylactic reactions (Kadivar & McClennan, 2010).
139
Risks and Complication of Coronary Angiography: Contrast Related Complications

Treatment of anaphylactic reactions is mostly supportive. In mild reactions anti histaminic


drugs are effective, but in more severe cases use of short acting steroids is mandatory and
even some critical care facilities may be necessary.

8. Alternatives to Contrast Media


Very high risk patients may be in need for other imaging techniques rather than using
contras media. One of these agents is Carbone dioxide which may be a good alternative to
iodinated CM. It may be used solely or in conjunction with low dose of iodinated CM. The
procedure is simple and the quality of images is good. With using modern technologies such
as digital subtract angiography the nephrotoxicity of this method is neglectable (Kessel etal.,
2002; Liss etal., 2005; Shaw & Kessel, 2006).
The other alternative is magnetic resonance angiography (MRA). Its main complication in
renal failure is nephrogenic systemic fibrosis (NSF). SO in patients with moderate to severe
renal failure it is contraindicated and in patients with mild renal failure its use is cautiously
recommended.

9. Conclusion
Contrast agents are associated with many complications. There are two more prevalent and
important complications related to contrast media: allergic reactions and CIN. The allergic
reactions are preventable by knowing the high risk patients and premedication with steroids
and antihistamines.
The most important complication of contrast agents is CIN, which is usually a mild to
moderate AKI that is rapidly recovered. But it could be associated with increased significant
morbidity and mortality. There is no treatment for developed CIN, so it should be
prevented by some measures. The proved modalities for prevention of CIN are saline
administration, the lowest possible dose of CM, and use of IOCM or LOCM.
All patients at risk should be evaluated carefully before the procedures with CM and the
risk factors should be stratified according to available risk stratification models.

10. References
Agmon Y, Peleg H, Greenfeld Z, etal. (1994) Nitric oxide and prostanoids protect the renal
outer medulla from radiocontrast toxicity in the rat. J Clin Invest; 94: 1069–1075.
Alonso A, Lau J, Jaber BL, et al. (2004) Prevention of radiocontrast nephropathy with N-
acetylcysteine in patients with chronic kidney disease: a meta-analysis of
randomized, controlled trials. Am J Kidney Dis; 43:1–9.
American Heart Association statistics committee and stroke subcommittee -2007 update: A
report from the AHA statistics committee and stroke subcommittee. Circulation
115:e69-e171.
Anto HR, Chou SY, Porush JG, Shapiro WB. (1981) Infusion intravenous pyelography and
renal function. Effect of hypertonic mannitol in patients with chronic renal
insufficiency. Arch Intern Med;141:1652–1656
Arakawa K, Suzuki H, Naitoh M, etal. (1996)Role of adenosine in the renal responses to
contrast medium. Kidney Int; 49:1199–1206.
140 Advances in the Diagnosis of Coronary Atherosclerosis

Araujo M, Welch WJ. (2006) Oxidative stress and nitric oxide in kidney function. Curr Opin
Nephrol Hypertens; 15:72–77.
Aspelin P, Aubry P, Fransson SG, etal. (2003)Nephrotoxic effects in high-risk patients
undergoing angiography. N Engl J Med;348:491–499.
Atkin JL. Effect of sodium bicarbonate preloading on ischemic renal failure. Nephron
1986;44:70–76.
Bader BD, Berger ED, Heede MB, et al. (2004) What is the best hydration regimen to prevent
contrast media-induced nephrotoxicity? Clin Nephrol; 62:1–7.
Bagnis C, Idee JM, Dubois M, et al. (1997) Role of endothelium-derived nitric oxide
endothelin balance in contrast medium-induced acute renal vasoconstriction in
dogs. Acad Radiol; 4: 343–348.
Bagshaw SM, Ghali WA. (2004) Acetylcysteine for prevention of contrast-induced
nephropathy after intravascular angiography: a systematic review and meta-
analysis. BMC Med; 2:38–49.
Bagshaw SM, Ghali WA. (2005) Theophylline for prevention of contrast-induced
nephropathy: a systematic review and meta analysis. Arch Intern Med; 165: 1087–
1093.
Bagshaw SM, McAlister FA, Manns BJ, Ghali WA. (2006)Acetylcysteine in the prevention of
contrast-induced nephropathy:A case study of the pitfalls in the evolution of
evidence. ArchIntern Med;166:161–166.
Bailey CJ, Turner RC. Metformin. N Engl J Med; 334:574–579.
Baker CS, Wragg A, Kumar S, etal. (2003) A rapid protocol for the prevention of
contrastinduced renal dysfunction: the RAPPID study. J Am Coll Cardiol; 41:2114–
2118.
Bakris GL, Burnett JC Jr. (1985) A role for calcium in radiocontrast induced reductions in
renal hemodynamics. Kidney Int; 27:465–468.
Bakris GL, Lass N, Gabaer AO, etal. (1990) Radiocontrast medium-induced declines in renal
function. A role for oxygen free radicals. Am J Physiol; 175:57–60.
Barrett BJ, Parfrey PS, Vavasour HM, et al. (1992) Contrast nephropathy in patients with
impaired renal function: high versus low osmolar media. Kidney Int;41: 1274– 1279.
Barrett B J,Carlisle E J. (1993) Meta-analysis of the relative nephrotoxicity of high- and low
osmolality iodinated contrast media . Radiology;188:171-178.
Barrett BJ, Katzberg RW, Thomsen HS, et al.( 2006) Contrast-induced nephropathy in
patients with chronic kidney disease undergoing computed tomography: A double
blind comparison of iodixanol and iopamidol. Invest Radiol;41:815–821.
Bartholomew BA, Harjai KJ, Dukkipati S, et al.(2004). Impact of nephropathy after
percutaneous coronary intervention and a method for risk stratification. Am J
Cardiol; 93:1515–1519.
Beeri R, Symon Z, Brezis M, et al. (1995) Rapid DNA fragmentation from hypoxia along the
thick ascending limb of rat kidneys. Kidney Int; 47: 1806–1810.
Bettmann MA. (2005)Contrast medium-induced nephropathy: critical review of the existing
clinical evidence. Nephrol Dial Transplant;20(suppl 1): i2–i7.
Birck R, Krzossok S, Markowetz F, et al. (2003) Acetylcysteine for prevention of contrast
nephropathy: meta-analysis. Lancet; 362:598–603.
141
Risks and Complication of Coronary Angiography: Contrast Related Complications

Boccolandro F, Amhad M, Smalling RW, Sdringola S. (2003) Oral acetylcysteine does not
protect renal function from moderate to high doses of intravenous radiographic
contrast. Cathet Cardiovasc Interv; 58:336–341.
Briguori C, Airoldi F, D’Andrea D, et al. (2007) Renal insufficiency following contrast media
administration trial (REMEDIAL): a randomized comparison of 3 preventive
strategies. Circulation; 115:1211–1217.
Briguori C, Colombo A, Airoldi F et al. (2005) Nephrotoxicity of low-osmolality versus iso-
osmolality contrast agents: impact of N-acetylcysteine. Kidney Int; 68:2250–2255.
Briguori C, Visconti G, Rivera N V, etal.( 2010) Cystatin C and contrast-induced acute
kidney injury. Circulation;121:2117-2122.
Carraro M, Malalan F, Antoinione R, et al.( 1998) Effects of a dimeric vs. a monomeric
nonionic contrast medium of renal function in patients with mild to moderate renal
insufficiency: a double-blind randomized clinical trial. Eur Radiol; 8:144– 147.
Cigarroa R G, Lange R A, Williams R H, Hillis L D. (1989) Dosing of contrast material to
prevent contrast nephropathy in patients with renal disease. Am J Med ;86: 649-652.
Cirit M, Toprak O, Yesil M, et al. (2006) Angiotensin-converting enzyme inhibitors as a risk
factor for contrast-induced nephropathy. Nephron Clin Pract; 104: c20- c27.
Costa N. (2004) Understanding contrast media. J Infus Nurs; 27:302–12.
Cruz D N, Perazella M A, Bellomo R, etal. (2006) Extracorporeal blood purification therapies
for prevention of radiocontrast-induced nephropathy: A systematic review. Am J
Kidney Dis; 48:361-371.
Dangas G, Iakovou I, Nikolsky E, et al. (2005) Contrast-induced nephropathy after
percutaneous coronary interventions in relation to chronic kidney disease and
hemodynamic variables. Am J Cardiol; 95:13–19.
Deray G. (2006) Dialysis and iodinated contrast media. Kidney Int Suppl;100:S25–S29.
Drager LF, Andrade L, Barros de Toledo JF, etal. (2004) Renal effects of N- acetylcysteine in
atients at risk for contrast nephropathy: decrease in oxidant stress-mediated
renaltubular injury. Nephrol Dial Transplant; 19:1803–1807.
Duong MH, MacKenzie TA, Malenka DJ. (2005) N-acetylcysteine prophylaxis
significantlyreduces the risk of radiocontrast-induced nephropathy: comprehensive
meta-analysis. Catheter Cardiovasc Interv; 64:471–479.
Dussol B, Morange S, Loundoun A, et al. (2006) A randomized trial of saline hydration to
prevent contrast nephropathy in chronic renal failure patients. Nephrol Dial
Transplant; 21:2120–2126.
Dzgoeva FU, Milovanov Iu S, Kutyrina IM. (1995) Acute kidney failure related to the use of
x-ray contrast agents and indomethacin: the risk factors and mechanisms of its
development. (Abstract)Ter Arkh; 67:36–39.
Efrati S, Dishy V, Averbukh M, etal. (2003) The effect of N-acetylcystein on renal function,
nitric oxide, and oxidative stress after angiography. Kidney Int; 64:2182–2187.
Erley C. (2006) Concomitant drugs with exposure to contrast media. Kidney Int; 69 Suppl
100:S20–S24.
Fishbane S. (2008) N-acetylcysteine in prevention of contrast-induced nephropathy.
Clin J Am Soc Nephrol; 3:281–287.
Frank H, Werner D, Lorusso V, et al. (2003) Simultaneous hemodialysis during coronary
angiography fails to prevent radiocontrast-induced nephropathy in chronic renal
failure. Clin Nephrol; 60:176–182.
142 Advances in the Diagnosis of Coronary Atherosclerosis

From AM, Bartholmai BJ, Williams AW, etal. (2008) Sodium bicarbonate is associated with
an increased incidence of contrast nephropathy: a retrospective cohort study of
7977 patients at mayo clinic. Clin J Am Soc Nephrol;3:10–18.
Gabutti L, Marone C, Monti M, etal. (2003) Does continuous veno-venous hemodiafiltration
concomitant with radiological procedures provide a significant and safe removal of
the iodinated contrast ioversol? Blood Purif; 21: 152–157.
Goldenberg I, Chonchol M, Guetta V.(2009)Reversible acute kidney injury followin contrast
exposure and the risk of long-term mortality. Am J Nephrol; 29:136–144.
Gruberg L, Mintz GGS, Mehran R, et al. (2000)The prognostic implications of further renal
function deterioration within 48 h of interventional coronary Procedur in patients
with preexistent chronic renal insufficiency. J Am Coll Cardiol; 36: 1542-1548.
Gruberg L,Mehran R,Dangas G, etal.( 2001) Acute renal failure requiring dialysis after
percutaneous coronary interventions. Catheter Cardiovasc Interv; 52:409- 416.
Gupta RK, Kapoor A, Tewari S, et al. (1996) Captopril for prevention of contrast induced
nephropathy in diabetic patients: a randomised study. Indian Heart J 1999; 51:521–
526.
Haller C, Hizoh I. (2004)The cytotoxicity of iodinated radiocontrast agents on renal cells in
vitro. Invest Radiol;39:149–154.
Hamani A, Petitclerc T, Jacobs C, Deray G. (1998) Is dialysis indicated immediately after
administration of iodinated contrast agents in patients on haemodialysis? Nephrol
Dial Transplant ;13:1051–1052.
Heinrich MC, Haberle L, Muller V, et al.( 2009) Nephrotoxicity of iso-osmolar Iodixanol
compared with nonionic low-osmolar contrast media: meta-analysis of randomized
controlled trials. Radiology; 250:68–86.
Heyman SN, Rosen S, Brezis M. (1994) Radiocontrast nephropathy: a paradigm for the
synergism between toxic and hypoxic insults in the kidney. Exp Nephrol; 2:153–157.
Heyman SN, Rosen S, Rosenberger C. (2008) Renal parenchymal hypoxia, hypoxi
adaptation, and the pathogenesis of radiocontrast nephropathy. Clin J Am Soc
Nephrol;3:288–296.
Hierholzer K, Hierholzer J: The discovery of renal contrast media in berlin. Am J
Nephrol 2002;22:295-299.
Hirsch R, Dent C, Pfriem H, etal.( 2007) NGAL is an early predictive biomarker of contrast-
induced nephropathy in children. Pediatr Nephrol;22:2089–2095.
Hoffmann U, Fischereder M, Kruger B, etal. (2004) The value of N-acetylcysteine in the
prevention of radiocontrast agent-induced nephropathy seems questionable. J Am
Soc Nephrol;15:407–410.
Hogan SE, L’Allier P, Chetcuti S, etal. (2008) Current role of sodium bicarbonate- based
preprocedural hydration for the prevention of contrast-induced acute kidney
injury: Am eta-analysis. Am Heart J; 156:414–421.
Hricak H, Barbariv Z L(1984).Newer imaging techniques:Workshop; in Robinson I (ed): Proc
9th Int Cong Nephrol, , 1430-1436.
Huber W, Eckel F, Hennig M,etal. (2006) Prophylaxis of contrast material-induced
nephropathy in patients in intensive care: acetylcysteine, theophylline, or both? A
randomized study. Radiology ;239:793–804.
Humes HD, Hunt DA, White MD. (1987) Direct toxic effect of the radiocontrast agent
diatrizoate on renal proximal tubule cells. Am J Physiol; 252: F246–255.Isenbarger
143
Risks and Complication of Coronary Angiography: Contrast Related Complications

DW, Kent SM, O’Malley PG. (2003) Meta-analysis of randomized clinical trials on
the usefulness of acetylcysteine for prevention of contrast nephropathy. Am J
Cardiol; 92:1454–1458.
Itoh Y, Yano T, Sendo T, etal. (2006) Involvement of de novo ceramide synthesis in
radiocontrast-induced renal tubular cell injury. Kidney Int; 69:288–297.
James MT, Ghali WA, Tonelli M, et al. (2010) Acute kidney injury following coronary
angiography associated with a long-term decline in kidney function. Kidney Int;
78:803-809.
Kadivar Kh, McClennan BL. (2010) Intravenous Iodinated Contrast Media Reactions:
Recognition and Response. Contemporary Diag Radiol ; 33:1-6.
Kanter M Z. (2006) Comparison of oral and i.v acetylcysteine in the treatment of
acetaminophen poisoning. Am J Health Syst Pharm; 63:1821–1827.
Kay J, Chow WH, Chan TM, etal. (2003) Acetylcysteine for prevention of acute deterioration
of renal function following elective coronary angiography and intervention: a
randomized controlled trial. JAMA; 289:553–558.
Kessel DO, Robertson I, Patel J etal. (2002) Carbon-dioxide-guided vascular interventions:
technique and pitfalls.Cardiovasc Intervent Radiol.;25(6):476.
Klarenbach SW, Pannu N, Tonelli MA, Manns BJ. (2006) Cost-effectiveness of hemofiltration
to prevent contrast nephropathy in patients with chronic kidney disease. Crit Care
Med;34:1044–1051.
Kshirsagar AV, Poole C, Mottl A, et al. (2004) N-acetylcysteine for the prevention of
radiocontrast induced nephropathy: a meta-analysis of prospective controlled
trials. J Am Soc Nephrol; 15:761–769.
Kushner FG, Hand M, Smith SC Jr, et al. (2009) focused updates: ACC/AHA guidelines for
the management of patients with ST-elevation myocardial infarction (updating the
2004 guideline and 2007 focused update) and ACC/AHA/SCAI guidelines on
percutaneous coronary intervention (updating the 2005 guideline and 2007 focused
update). A report of the American College of Cardiology Foundation/American
Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol; 54:2205–
2241.
Laskey WK, Jenkins C, Selzer F, et al. (2007)Volume-to-creatinine clearance ratio. J Am Coll
Cardiol; 50:584–590.
Lee PT, Chou KJ, Liu CP, etal. (2007) Renal protection for coronary angiography in
advanced renal failure patients by prophylactic hemodialysis. A randomized
controlled trial. J Am Coll Cardiol;50:1015–1020.
Liss P, Nygren A, Erikson U, Ulfendahl HR. (1998) Injection of low and iso-osmolar contrast
medium decreases oxygen tension in the renal medulla. Kidney Int;53:698–702.
Liss P, Carlsson PO, Palm F, Hansell P. (2004) Adenosine A1 receptors in contrast media-
induced renal dysfunction in the normal rat. Eur Radiol;14:1297–1302.
Liss P, Eklöf H, Hellberg O, etal. (2005) Renal effects of CO2 and iodinated contrast media in
patients undergoing renovascular intervention: a prospective, randomized study. J
Vasc Interv Radiol.;16(1):57.
Liss P, Persson PB, Hansell P, Lagerqvist B. (2006) Renal failure in 57 925 patients
undergoing coronary procedures using isosmolar or low-osmolar contrast media.
Kidney Int ;70:1811–1817.
144 Advances in the Diagnosis of Coronary Atherosclerosis

Liu R, Nair D, Ix J, et al. (2005) N-acetylcysteine for the prevention of contrast- induced
nephropathy: a systematic review and meta-analysis. J Gen Intern Med;20:193–200.
Manske CL, Sprafka JM, Strony JT, Wang Y. (1990) Contrast nephropathy in azotemic
diabetic patients undergoing coronary angiography. Am J Med; 89:615–620.
Maioli M, Toso A, Leoncini M, etal. (2008) Sodium bicarbonate versus Saline for the
prevention of contrast-induced nephropathy in Patients with renal dysfunction
undergoing coronary angiography or intervention. J Am Coll Cardiol;52:599–604.
Marenzi G, Marana I, Lauri G, et al. (2003) The prevention of radio contrast- agent induced
nephropathy by hemofiltration. N Engl J Med; 349:1333–1340.
Marenzi G, Lauri G, Assanelli E, et al. (2004) Contrast-induced nephropathy in patients
undergoing primary angioplasty for acute myocardial infarction. J Am Coll Cardiol;
44:1780–1785.
Marenzi G, Lauri G, Campodonico J, et al. (2006b) Comparison of two hemofiltration
protocols for prevention of contrast-induced nephropathy in high –risk patients.
Am J Med; 119:155-162
Marenzi G, Assanelli E, Marana I, etal. (2006a) N-acetylcysteine and contrast-induced
nephropathy in primary angioplasty. N Engl J Med;354:2773–2782.
Marenzi G, Assanelli E, Campodonico J, etal. (2009) Contrast volume during primary
percutaneous coronary intervention and subsequent contrast-induced nephropathy
and mortality. Ann Intern Med; 150:170–177.
Masuda M, Yamada T, Mine T, et al. (2007) Comparison of usefulness of sodium bicarbonate
versus sodium chloride to prevent contrast-induce nephropathy in patients
undergoing emergent coronary procedure. Am J Cardiol; 100:781–786.
McCullough PA, Wolyn R, Rocher LL, et al. (1997) Acute renal failure after coronary
intervention: incidence, risk factors, and relationship to mortality. Am J Med;
103:368–375.
McCullough P A, Soman S S. (2005) Contrast-induced nephropathy. Crit Care Clin; 21:261–
280.
McCullough PA, Adam A, Becker CR, etal. (2006a) Epidemiology and prognostic
implications of contrast-induced nephropathy. Am J Cardiol;98:5K–13K.
McCullough PA, Adam A, Becker CR, et al. (2006b) Risk prediction of contras induced
nephropathy. Am J Cardiol; 98:27K–36K.
McCullough PA, Bertrand ME, Brinker JA, Stacul F. (2006c) A meta-analysis of the renal
safety of isosmolar iodixanol compared with low-osmolar contrast media. J Am Coll
Cardiol; 48:692–699.
Mehran R, Aymong ED, Nikolsky E, et al. (2004) A simple risk score for prediction of
contrast-induced nephropathy after percutaneous coronary intervention:
development and initial validation. J Am Coll Cardiol; 44:1393–1399.
Merten GJ, Burgess WP, Gray LV, et al. (2004) Prevention of contrast-induced nephropathy
with sodium bicarbonate: a randomized controlled trial. JAMA; 291:2328–2334.
Mockel M, Radovic M, Kuhnle Y, etal. (2008) Acute renal haemodynamic effects of
radiocontrast media in patients undergoing left ventricular and coronary
angiography. Nephrol Dial Transplant;23:1588–1594
Morcos S K, Thomsen H S, Webb J A W and members of contrast media safety committee of
the European Society of Urogenital Radiology (ESUR). (1999) Contrast media
induced nephropathy: a consensus report. Eur Radiol;9:1602-1613.
145
Risks and Complication of Coronary Angiography: Contrast Related Complications

Morcos SK, Thomsen HS, Webb JA. (2002) Dialysis and contrast media. Eur Radiol; 12: 3026–
3030.
Morcos SK. (2005) Prevention of contrast media-induced nephrotoxicity after angiographic
procedures. J Vasc Interv Radiol;16:13–23.
Mueller C, Buerkle G, Buettner HJ, et al. (2002) Prevention of contrast media associated
nephropathy: randomized comparison of 2 hydration regimens in 1620 patients
undergoing coronary angioplasty. Arch Intern Med; 162:329–336.
Nallamothu BK, Shojania KG, Saint S, et al. (2004) Is acetylcysteine effective in preventing
contrast-related nephropathy? A meta-analysis. Am J Med; 117:938–947.
Nash K, Hafeez A, Hou S. (2002) Hospital-acquired renal insufficiency. Am J Kidney Dis;
39:930–936.
Nikolsky E, Mehran R, Lasic Z, etal. (2005) Low hematocrit predicts contrast-induced
nephropathy after percutaneous coronary interventions. Kidney Int;67:706–713.
Onbasili AO, Yenicerioglu Y, Agaoglu P, et al. (2007) Trimetazidine in the prevention of
contrast-induced nephropathy after coronary procedures. Heart;93:654-655.
Ozcan EA, Guneri S, Akdeniz B, et al. (2007) Sodium bicarbonate, N acetylcysteine, and
saline for prevention of radio-contrast-induced nephropathy. A comparison of 3
regimens for protecting contrast-induced nephropathy in patients undergoing
coronary procedures. A single-center prospective controlled trial. Am Heart J;
154:539–544.
Pannu N, Manns B, Lee H, Tonelli M. (2004) Systematic review of the impact of N-
acetylcysteine on contrast nephropathy. Kidney Int; 65:1366–1374
Peer A, Averbukh Z, Berman S, etal.(2003) Contrast media augmented apoptosis of cultured
renal mesangial, tubular, epithelial, endothelial, and hepatic cells. Invest Radiol;
38:177–182
PerazellaMA.(2009)Radiocontrast-induced nephropathy: an update. Minerva Urol Nefrol ;
61:215-233.
Persson PB (2005).Contrast medium-induced nephropathy. Nephrol Dial Transplant; 20 Suppl
1:i1
Persson PB, Tepel M.(2006) Contrast medium-induced nephropathy: the pathophysiology.
Kidney Int Suppl; 100:S8–10.
Pflueger A, Larson TS, Nath KA, et al. (2000) Role of adenosine in contrast media induced
acute renal failure in diabetes mellitus. Mayo Clin Proc; 75:1275–1283.
Ranucci M, Ballotta A, Kunkl A, et al.( 2008)Influence of the timing of cardiac catheterization
and the amount of contrast media on acute renal failure after cardiac surgery. Am J
Cardiol; 101:1112–1118.
Recio-Mayoral A, Chaparro M, Prado B, et al. (2007) The reno-protective effect of hydration
with sodium bicarbonate plus N-acetylcysteine in patients undergoing emergency
percutaneous coronary intervention: the RENO study. J Am Coll Cardiol; 49:1283–
1288.
Reed M, Meier P, Tamhane U U, etal.( 2009)The relative safety of Iodixanol compared with
low-osmolar contrast media: a meta-analysis of randomized controlled trials. J Am
Coll Cardiol;2:645-654.
Ribeiro L, de Assuncao e Silva F, Kurihara RS, etal. (2004) Evaluation of the nitric oxide
production in rat renal artery smooth muscle cells culture exposed to radiocontrast
agents. Kidney Int; 65: 589–96.
146 Advances in the Diagnosis of Coronary Atherosclerosis

Ribichini F, Graziani M, Gambaro G, etal. (2010) Early Creatinine shifts Predict Contrast-
induced Nephropathy and persistent renal damage after angiography. Am J Med;
123:755-763.
Rihal CS, Textor SC, Grill DE, et al. (2002) Incidence and prognostic importance of acute
renal failure after percutaneous coronary intervention. Circulation; 105:2259–2264.
Rudnick M R,Goldfarb S,Wexler L,etal. (1995)Nephrotoxicity of ionic and nonionic contrast
media in 1196 patients: A randomized trial: the Iohexol Cooperative Study.
KidneyInt;47:254-261.
Rudnick M, Kesselheim A, Goldfarb S. (2006) Contrast-induced nephropathy: how it
develops, how to prevent it. Cleveland Clin J Med; 73:75–87.
Rudnick M, Goldfarb S. (2003) Pathogenesis of contrast-induced nephropathy: Experimental
and clinical observations with an emphasis on the role of osmolality. Rev Cardiovasc
ed; 4 (Suppl 5):S28–S33.
Rudnick MR, Davidson C, Laskey W, et al.( 2008) Nephrotoxicity of iodixanol versus
ioversol in patients with chronic renal disease. The visipaque angiography
/interventions with laboratory outcomes in renal insufficiency (VALOR) trial.
Am Heart J;156:776-782.
Rudnick M, Feldman H. (2008) Contrast-induced nephropathy: what are the true clinical
consequences? Clin J Am Soc Nephrol; 3:263–272.
Sayol A J, Bush W H. (2011) Avoidable errors in dealing with anaphylactoid reactions to
iodinated contrast media. Invest Radiol;46: 147–151.
Seeliger E, Flemming B, Wronski T, et al.( 2007) Viscosity of contrast media perturbs renal
hemodynamics. J Am Soc Nephrol; 18:2912–2920.
Shah SH, Mehta RL. Acute kidney injury in critical care:time for a paradigm shift? Curr Opin
Nephrol Hypertens 2006;15:561– 565.
Shaw DR, Kessel DO. (2006) The current status of the use of carbon dioxide in diagnostic
and interventional angiographic procedures. Cardiovasc Intervent Radiol. ;29(3):323.
Solomon R, Werner C, Mann D, et al. (1994) Effects of saline, mannitol, and furosemide to
prevent acute decreases in renal function induced by radiocontrast agents. N Engl J
Med; 331:1416–1420.
Solomon R. (2005)The role of osmolality in the incidence of contrast induced nephropathy:
A systematic review of angiographic contrast media in high risk patients. Kidney
Int;68:2256–2263.
Solomon R, Dumouchel W. ( 2006) Contrast media and nephropathy: findings from
systematic analysis and Food and Drug Administration reports of adverse effects.
Invest Radiol; 41:651-660.
Solomon R , Natarajan MK, Doucet S, et al. (2007) Cardiac angiography in renally impaired
patients (CARE) study: A randomized double-blind trial of contrast induced
nephropathy in patients with chronic kidney disease. Circulation; 115:3189–3196.
Solomon RJ, Mehran R, Natarajan MK, et al. (2009) Contrast-induced nephropathy and
long-term adverse events: cause and effect? Clin J Am Soc Nephrol; 4:1162–1169.
Spargias K, Alexopoulos E, Kyrzopoulos S, et al. (2004) Ascorbic acid prevents contrast-
mediated nephropathy in patients with renal dysfunction undergoing coronary
angiography or intervention. Circulation; 110:2837–2842.
Stevens MA, McCullough PA, Tobin KJ, et al. (1999) A prospective randomized trial of
prevention measures in patients at high risk for contrast nephropathy: results of the
147
Risks and Complication of Coronary Angiography: Contrast Related Complications

P.R.I.N.C.E. study. Prevention of radiocontrast induced nephropathy clinical


evaluation. J Am Coll Cardiol; 33:403–411.
Subramanian S, Tumlin J, Bapat B, Zyczynski T. (2007) Economic burden of contrast-
induced nephropathy: implications for prevention strategies. J Med Econ; 10:119–
134.
Taylor AJ, Hotchkiss D, Morse RW, McCabe J. (1998) PREPARED: Preparation for
angiography in Renal Dysfunction: a randomized trial of inpatient vs. outpatient
hydration protocols for cardiac catheterization in mild-to-moderate renal
dysfunction. Chest; 114:1570–1574.
Tepel M, van der Giet M, Schwarzfeld C, et al. (2000) Prevention of radiographic contrast-
agent-induced reductions in renal function by acetylcysteine. N EnglJ Med;
343:180–184.
Tepel M, Aspelin P, Lameire N. (2006) Contrast-induced nephropathy: a clinical and
evidence-based approach. Circulation;113:1799–806.
The Committee on Drugs and Contrast Media of the American College of Radiology (2010).
Contrast nephropathy. In: ACR manual oncontrast media,Version 7.28 American
College of Radiology, Retrieved from: http://www.acr.org/
secondarymainmenucategories/quality_safety/contrast_manual/fullmanual.aspx
Thomsen H S. (2005)How to avoid CIN: guidelines from the European Society of Urogenital
Radiology. Nephrol Dial Transplant; 20 (Suppl 1):i18– i22.
Thomsen H S, Morcos S K, Members of Contrast Media Safety Committee of European
Society of Urogenital Radiology (ESUR). (2005) In which patients should serum
creatinine be measured before contrast medium administration? Eur Radiol, 15:
749–756.
Thomsen H S, Morcos SK. (2006) ESUR guidelines on contrast media. Abdom Imaging; 31:
131–140.
Thomsen HS. (2006) Reducing the risk of contrast media induced nephrotoxicity. In:
Contrast media: safety issues and ESUR Guidelines. Thomsen HS (ed) 53–55
Springer, 978-3-540-72783-5 Heidelberg Berlin New York,; pp.
Thomsen HS, Morcos SK, Erley CM, et al. (2008)The ACTIVE trial: comparison of the effects
on renal function of iomeprol-400 and iodixanol-320 in patients with chronic
kidney disease undergoing abdominal computed tomography. Invest Radiol;
43:170–178.
Toprak O, Cirit M, Bayata S, etal. (2003) The effect of preprocedural captopril on contrast-
induced nephropathy in patients who underwent coronary angiography.
(abstract) Anadolu Kardiyol Derg;3:98-103.
Toprak O, Cirit M.(2006) Risk factors and therapy strategies for contrast-
inducednephropathy. Renal Fail;28:365–381.
Trivedi HS, Moore H, Nasr S, et al. (2003) A randomized prospective trial to assess the role
of saline hydration on the development of contrast nephrotoxicity. Nephron Clin
Pract; 93:c29–c34.
Ultramari FT, Bueno Rda R, da Cunha CL, et al.(2006)Contrast media-induced nephropathy
following diagnostic and therapeutic cardiac catheterization. Arq bras Cardiol;
87:378–90.
Vasheghani -Farahani A, Sadigh G, Kassaian S E etal. (2009)Sodium Bicarbonate Plus
Isotonic Saline Versus Saline for Prevention of contrast-induced nephropathy in
148 Advances in the Diagnosis of Coronary Atherosclerosis

patients undergoing coronary angiography: A randomized controlled trial; Am J


Kidney Dis 54:610-618.
Vasheghani-Farahani A ,Sadigh G ,Kassaian S, etal. (2010) Sodium bicarbonate in
preventing contrast nephropathy in patients at risk for volume overload: a
randomized controlled trial. J Nephrol; 23:216–223.
Vogt B, Ferrari P, Schonholzer C, et al. (2001) Prophylactic hemodialysis after radiocontrast
media in patients with renal insufficiency is potentially harmful. Am J Med; 111:
692–698.
Weinmann H J, Platzec J, Schirmer H etal.(2005) Contrast media: future aspects. Eur Radiol15
(Suppl4):D70-D73
Weisberg LS, Kurnik PB, Kurnik BR (1992) Radiocontrastinduced nephropathy in humans:
role of renal vasoconstriction. Kidney Int 41:1408–1415.
Weisberg LS, Kurnik PB, Kurnik BR. (1994) Risk of radiocontrast nephropathy in patients
with and without diabetes mellitus. Kidney Int;45:259–265.
Weisbord SD, Chen H, Stone RA, etal.(2006)Associations of increases in serum Creatinine
with mortality and length of hospital stay after coronary angiography. J Am Soc
Nephrol ; 17:2871–2877.
Weisbord SD, Pavelsky PM. (2008) Prevention of contrast-induced nephropathy with
volume expansion. Clin J Am Soc Nephrol; 3:273–280.
Yokomaku Y, Sugimoto T, Kume S, et al. (2008) Asialoerythropoietin prevents contrast-
induced nephropathy. J Am Soc Nephrol; 19:321–328.
Zagler A, Azadpour M, Mercado C, Hennekens CH. (2006) N-acetylcysteine and contrast-
induced nephropathy: a meta-analysis of 13 randomized trials. Am Heart J;
151:140–145.
8

Complications of Cardiac
Catetherization
Mariano García-Borbolla1, Rafael García-Borbolla2
and Begoña Balboa3
1Hospital Virgen Macarena
2Hospital
de Jerez
3Hospital Virgen del Rocio

Spain

1. Introduction
Cardiac catheterization was primarily a diagnostic procedure that was used to evaluate
hemodynamics, ventricular function, and coronary anatomy. However, the introduction of
improved angioplasty equipment and new interventional devices in the 1980s made
catheterization an important tool in the treatment of cardiovascular disease.
Although diagnostic catheterization and percutaneous coronary intervention are done by
skin puncture rather than incision, and under local rather than general anesthesia, they can
cause a variety of adverse events, ranging from minor problems without long-term sequelae
to major complications requiring immediate corrective action. Furthermore, newer
interventional procedures may be associated with different and, in some cases, more
frequent complications.

2. Major complications
The risk of producing a major complication (death, myocardial infarction, or major
embolization) during diagnostic cardiac catheterization is generally well below 1 percent. As
a result, the risk-to-benefit ratio still favors performing this procedure as a part of the
evaluation of potentially fatal or lifestyle limiting cardiac disease.
The risk of an adverse event, for the individual patient, is dependent upon demographics,
cardiovascular anatomy, clinical circumstances including comorbidities, the experience of
the operator, and the type of procedure being performed (table 1 A-C) [1]. Severe peripheral
artery disease is a risk factor for all of the major complications.

2.1 Death
Death following diagnostic cardiac catheterization is now rare. Compared with the 1 percent
mortality rate seen in the 1960s, the second registry of the Society for Cardiac Angiography
revealed that the procedural mortality had fallen to 0.1 percent in the more than 200,000
patients catheterized between 1984 and 1987 [2].
High-risk subgroups have been identified in multiple large series [1-9]:
150 Advances in the Diagnosis of Coronary Atherosclerosis

Percent
Mortality 0.11
Myocardial infarction 0.05
Cerebrovascular accident 0.07
Arrhythmia 0.38
Vascular complications 0.43
Contrast reaction 0.37
Hemodynamic complications 0.26
Perforation of heart chamber 0.28
Other complications 0.28
Total of major complications 1.70
Noto, TJ Jr, Johnson, LW, Krone, R, et al. Cardiac catheterization 1990: A report of the Registry of the
Society for Cardiac Angiography and Interventions (SCA&I). Cathet Cardiovasc Diagn 1991; 24:75.
Table 1. (A): Risk of cardiac catheterization and coronary angiography (based on 59,792
patients)

Variable Odds ratio


Moribund 10.22
Shock 6.52
Acute MI <24 h 4.03
Renal insufficiency 3.30
Cardiomyopathy 3.29
Aortic valve disease 2.72
Mitral valve disease 2.33
Heart failure 2.33
New York Heart Association Functional
Class I 1.00
Class II 1.15
Class III 1.32
Class IV 1.52
Hypertension 1.45
Unstable angina 1.42
Outpatient/inpatient 0.63
Moribund indicates a patient who responds poorly due to a life threatening condition.
MI: myocardial infarction.
Scanlon, PJ, Faxon, DP, Audet, AM, et al, J Am Coll Cardiol 1999; 33:1756.
Table 1. (B): Multivariate predictors of major complications of coronary angiography (based
on 58,332 procedures)
Complications of Cardiac Catetherization 151

Acute renal failure


Chronic renal failure secondary to diabetes
Active gastrointestinal bleeding
Unexplained fever, which may be due to infection
Untreated active infection
Acute stroke
Severe anemia
Severe uncontrolled hypertension
Severe symptomatic electrolyte imbalance
Severe lack of cooperation by patient due to psychological or severe systemic illness
Severe concomitant illness that drastically shortens life expectancy or increases risk of
therapeutic interventions
Refusal of patient to consider definitive therapy such as PTCA, CABG, or valve
replacement
Digitalis intoxication
Documented anaphylactoid reaction to angiographic contrast media
Severe peripheral vascular disease limiting vascular access
Decompensated heart failure or acute pulmonary edema
Severe coagulopathy
Aortic valve endocarditis

PTCA: percutaneoas transluminal coronary angioplasty; CABG: coronary artety bypass graft.
Scanlon, PJ, Faxon, DP, Audet, AM, et al, J Am Coll Cardiol 1999; 33:1756.
Table 1. (C): Relative contraindications to coronary angiography

 Patients with valvular heart disease, chronic kidney disease, diabetes mellitus requiring
insulin therapy, peripheral artery disease, cerebrovascular disease, and pulmonary
insufficiency all appear to have an increased incidence of death and major
complications from cardiac catheterization. Mortality is especially high in those with
preexisting renal insufficiency who have further deterioration of renal function within
48 hours after the procedure, particularly when dialysis is required [10].
 A similar increase in risk is seen with severe disease of the left main coronary artery
(compared to single vessel disease) and with a left ventricular ejection fraction (LVEF)
below 30 percent (compared to an LVEF above 50 percent).
 Age above 60 years and below one year and women have an increased mortality rate
after catheterization.
 New York Heart Association functional class IV is associated with an almost tenfold
increase in mortality compared with classes I and II (table 2).
152 Advances in the Diagnosis of Coronary Atherosclerosis

New York Heart


Canadian Cardiovascular Society
Class Association functional Specific activity scale
functional classification
classification
Patients can perform to
Patients with cardiac completion any activity
disease but without Ordinary physical activity, such requiring ≥7 metabolic
resulting limitations of as walking and climbing stairs, equivalents, eg, can carry
physical activity. Ordinary does not cause angina. Angina 24 lb up eight steps; do
I
physical activity does not with strenuous or rapid outdoor work (shovel
cause undue fatigue, prolonged exertion at work or snow, spade soil); do
palpitation, dyspnea, or recreation. recreational activities
anginal pain. (skiing, basketball, squash,
handball, jog/walk 5 mph).
Patients can perform to
Slight limitation of ordinary
completion any activity
activity. Walking or climbing
Patients with cardiac requiring ≤ 5 metabolic
stairs rapidly, walking uphill,
disease resulting in slight equivalents, eg, have
walking or stair climbing after
limitation of physical sexual intercourse without
meals, in cold, in wind, or when
activity. They are stopping, garden, rake,
under emotional stress, or only
II comfortable at rest. weed, roller skate, dance
during the few hours after
Ordinary physical activity fox trot, walk at 4 mph on
awakening. Walking more than
results in fatigue, level ground, but cannot
two blocks on the level and
palpitation, dyspnea, or and do not perform to
climbing more than one flight of
anginal pain. completion activities
ordinary stairs at a normal pace
requiring ≥ 7 metabolic
and in normal conditions.
equivalents.
Patients can perform to
completion any activity
Patients with cardiac requiring ≤ 2 metabolic
disease resulting in equivalents, eg, shower
marked limitation of Marked limitation of ordinary without stopping, strip and
physical activity. They are physical activity. Walking one to make bed, clean windows,
III comfortable at rest. Less two blocks on the level and walk 2.5 mph, bowl, play
than ordinary physical climbing more than one flight in golf, dress without
activity causes fatigue, normal conditions. stopping, but cannot and
palpitation, dyspnea, or do not perform to
anginal pain. completion any activities
requiring > 5 metabolic
equivalents.
Patient with cardiac
disease resulting in
inability to carry on any Patients cannot or do not
physical activity without perform to completion
Inability to carry on any physical
discomfort. Symptoms of activities requiring > 2
activity without discomfort -
IV cardiac insufficiency or of metabolic equivalents.
anginal syndrome may be present
the anginal syndrome may Cannot carry out activities
at rest.
be present even at rest. If listed above (Specific
any physical activity is activity scale III).
undertaken, discomfort is
increased.
Table 2. Comparison of three methods of assessing cardiovascular disability
Complications of Cardiac Catetherization 153

2.2 Myocardial infarction


Although periprocedural myocardial ischemia is fairly common, infarction is rare. The
general risk of myocardial infarction (MI) in most series is less than 0.1 percent [2-7].
However, the risk of precipitating an MI in an individual patient is influenced by patient-
related and technique-related factors. Risk factors that predispose patients to MI during the
procedure include:
 Recent non-ST elevation acute coronary syndrome.
 Extension of disease (0.17 percent with left main coronary disease versus 0.06 percent in
single vessel disease) [2].
 Diabetes mellitus requiring insulin therapy [1].

2.3 Stroke and transient ischemic attack


A stroke is a rare but potentially devastating complication of cardiac catheterization. The
incidence of stroke was 0.07 to 0.10 percent in an analysis of two large databases [2,11] and
has more recently been reported to be as high as 0.2 to 0.4 percent [12]. Risk factors for
stroke include the severity of coronary artery disease, the length of fluoroscopy time,
diabetes, hypertension, prior stroke, or renal failure [11,12]
Stroke and transient ischemic attacks are thought to be caused by disruption of
atheromatous plaques on the walls of the aorta. The risk of intracerebral hemorrhage is
increased in those receiving anticoagulation or thrombolytic therapy as well as with any of
the following: age ≥75 years; female sex; systolic blood pressure ≥160 mmHg; black race; and
low body weight (table 3). Based on the number of the risk factors present, the rate of
intracerebral hemorrhage varies from less than 1 percent to more than 4 percent.

Risk Factors*
Age ≥75 years
Black race
Female sex
Prior history of stroke
Systolic blood pressure ≥160 mmHg
Weight ≤65 kg for women or ≤80 kg for men
INR >4 or PT >24•
Use of alteplase (versus other thrombolytic agent)
Risk score Rate of intracranial hemorrhage, percent
0 or 1 0.69
2 1.02
3 1.63
4 2.49
≥5 4.11
* Each risk factor is worth 1 point if present, 0 points if absent. Points are added to determine the risk
score.
• INR: international normalized ratio; PT: prothrombin time.
Brass, LM, Lichtman, JH, Wang, Y, et al. Intracranial hemorrhage associated with thrombolytic therapy
for elderly patients with acute myocardial infarction: results from the cooperative cardiovascular
project. Stroke 2000; 31:1802.
Table 3. Cooperative cardiovascular project risk model for intracranial hemorrhage with
thrombolytic therapy
154 Advances in the Diagnosis of Coronary Atherosclerosis

Meticulous attention to technical factors such as wire and catheter exchanges is mandatory
in all patients, regardless of risk. Transient neurologic deficits may also result from the
injection of high osmolar contrast agents into the carotid or vertebral arteries. The majority
of patients with a periprocedural stroke have an unfavorable neurologic outcome and, in
one study, the in-hospital mortality was 32 percent [11].

2.4 Role of valvular aortic stenosis


The risk of stroke may be particularly high in patients with valvular aortic stenosis (AS) who
undergo retrograde catheterization of the aortic valve [13,14]. This was demonstrated in a
study of 152 patients with severe AS (mean age 71 years) who were randomly assigned to
cardiac catheterization with or without catheter passage through the valve [13]. The
following findings were noted:
 Detailed neurologic examination done before and after the catheterization
demonstrated clinically apparent deficits in 3 percent of those who underwent
retrograde catheterization, but in none of the other patients.
 Cranial MRI done before and after the catheterization demonstrated focal lesions
consistent with cerebral emboli in 22 percent of those who underwent retrograde
catheterization of the aortic valve, but in none of the patients who did not.
As a result, retrograde catheterization should be performed with caution in patients with
severe calcific AS and only when the information sought cannot be reliably obtained
noninvasively [14].

2.5 Asymptomatic embolic events


The incidence of stroke after cardiac catheterization was 0.07 to 0.10 percent in an analysis of
two large databases [2,11]. However, asymptomatic embolic events are much more common
[13,15,16].
This was illustrated in a prospective study of 48 patients (mean age 62 years) who
underwent cardiac catheterization and angiography and, in some patients, percutaneous
coronary intervention; magnetic resonance imaging (MRI) was performed before and about
24 hours after the procedure [15]. There were no embolic lesions on diffusionweighted
imaging at baseline, but seven patients (15 percent) had nine cerebral infarcts after the
procedure; all were asymptomatic.
Similar findings were noted in a report of 47 unselected patients in whom transcranial
Doppler was used to detect microemboli and MRI to detect new lesions [16]. The median
number of solid (ie, not gaseous) microemboli was significantly higher with a transradial
compared to a transfemoral access (57 versus 36). New MRI lesions occurred in 5 of 33
patients (15 percent) after transradial catheterization, compared to none of nine after
transfemoral catheterization. Most of the patients with new lesions remained asymptomatic.
These observations suggest that catheter manipulation in the diseased aortic root releases
small pieces of atherosclerotic debris more commonly than suspected, based upon the
fortunately low incidence of clinically apparent stroke. The same principles presumably
apply to peripheral embolism.

3. Local vascular complications


Local complications at the site of catheter insertion are among the most common problems
seen after cardiac catheterization.
Complications of Cardiac Catetherization 155

These problems may include acute thrombosis, distal embolization, dissection or poorly
controlled bleeding (which may be manifested as free hemorrhage or contained hematoma
in the femoral or retroperitoneal areas), pseudoaneurysm, or arteriovenous fistula.
Hemorrhage and hematoma are usually evident within 12 hours after the procedure by
virtue of local discomfort, hypotension, or falling hematocrit, which lead to confirmatory
noninvasive testing (ultrasound or CT). In contrast, other complications such as
pseudoaneurysm and arteriovenous fistula may not become apparent for days to weeks
afterward.

3.1 Hemostasis at the access site


The traditional approach to catheter removal at the conclusion of cardiac catheterization
was local hand-applied pressure for 15 to 30 minutes.
Two alternative approaches are available: mechanical clamp compression and more
importantly, arterial puncture closing devices:

3.1.1 Mechanical clamp


Mechanical clamp compression reduces the amount of staff time and effort required to
assure hemostasis, and may reduce the rate of vascular complications. One trial randomly
assigned 778 patients to hand applied pressure or mechanical clamp pressure; the latter
reduced the frequency of ultrasound-defined femoral vascular complications (femoral artery
thrombosis, echogenic hematoma, pseudoaneurysm, or arteriovenous fistulae formation) by
63 percent [17]. However, clamping does not decrease the importance of having staff in
attendance to ensure that the device is placed and remains in the correct position.
The circumstances which may make mechanical clamps the preferred method of achieving
hemostasis include procedures where longer hemostasis time is anticipated: larger sheaths,
excessively anticoagulated patients, recognized hematoma, and dual antiplatelet therapy

3.1.2 Closure devices


Arterial puncture closing devices (APCDs) eliminate the need for prolonged arterial
compression and reduce the subsequent interval of bed rest that is otherwise required. Two
types of APCDs in clinical use include collagen hemostatic puncture closure devices [18-21]
and suture-mediated devices [22].
APCDs may increase the risk of local vascular complications. In a meta-analysis of 30 trials
including 4000 patients who underwent coronary angiography or PCI, nonsignificant
increases in the incidence of groin hematoma, bleeding, and pseudoaneurysm formation
were associated with use of these devices compared to manual compression [23]. When only
those trials with an intention-to-treat analysis were included, the relative risks of groin
hematoma and pseudoaneurysm formation were significantly increased (1.89 and 5.40,
respectively).
A larger meta-analysis that included 30 studies involving 37,066 patients concluded that the
risk of complications depended upon the specific device used and whether the procedure
was a diagnostic catheterization or a PCI [24]. In the overall analysis, there was a significant
increase in risk with an APCD (odds ratio [OR] 1.34, 95% CI 1.01- 1.79). For reports limited
to diagnostic studies, there were no significant differences between APCDs and mechanical
compression, although there were trends toward fewer complications with the AngioSeal
156 Advances in the Diagnosis of Coronary Atherosclerosis

device (OR 0.71) and toward more complications with the VasoSeal and Perclose devices
(OR 1.85 and 1.51, respectively).
For reports limited to PCI, there were no significant differences between APCDs and
mechanical compression overall. However, there was a significant increase in vascular
complications with the VasoSeal device (OR 2.52).
Closure devices also increase the risk of local infection or endarteritis [25,26]. Although this
complication is infrequent (0.3 percent in one series) [26], it can be serious when it occurs.
Consideration should be given to periprocedural antibiotics in immunocompromised
patients following prolonged arterial access or when sterility has been breached when a
closure device is used. It may also be prudent for the operator to change gloves prior to
using an arterial closure device. (See 'Infection' below.) An angiogram of the femoral artery
should be performed prior to use of an APCD. Use ofclosure devices should be avoided if a
hematoma has formed during the procedure, or in the presence of severe iliofemoral
vascular disease, and when the access site is below the bifurcation of the superficial and
profunda femoral arteries.

3.1.3 Prior warfarin therapy


Among patients who are being treated with warfarin, the optimal approach is to withhold
warfarin and wait until the INR reaches 1.5 before proceeding with catheterization. When
the procedure cannot be delayed, it has been suggested that, in patients with an INR of 2.0
to 3.0, manual pressure hemostasis may be sufficient to prevent excess bleeding [27]. In
patients with an INR >3.0, administration of fresh frozen plasma prior to arterial sheath
removal may be warranted. The transradial approach may also be useful in such cases. The
risk of local bleeding in warfarin-treated patients is likely to be greater in those undergoing
PCI who are often treated with heparin, clopidogrel, and a glycoprotein IIb/IIIa inhibitor.
These patients typically have longer intraarterial times and more manipulation of the
arterial access site.increase in risk is seen with severe disease of the left main coronary artery
(compared to single vessel disease) and with a left ventricular ejection fraction (LVEF) below
30 percent (compared to an LVEF above 50 percent).

3.2 Hematoma formation


Hematoma formation is not uncommon in the soft tissue of the upper thigh and the patient
should be advised that the development of a large area of ecchymosis is likely. Although
most resolve over a period of days, femoral nerve compression can occur, which may take
weeks to months to resolve. Patients should be advised to avoid strenuous activity or lifting
heavy objects within the first week following arterial catheterization. Surgical repair for a
hematoma is generally not required.

3.2.1 Retroperitoneal extension


If the arterial puncture occurs above the inguinal ligament, hematoma may extend into the
retroperitoneal space, causing hypotension and ipsilateral flank pain. CT scanning or
abdominal ultrasonography can help to establish the diagnosis. Treatment is usually
conservative, consisting of bed rest and blood transfusion rather than surgical correction.
Reversal of systemic anticoagulation may be indicated if bleeding persists or is associated
with hemodynamic compromise.
Complications of Cardiac Catetherization 157

3.3 Arteriovenous fistula


Ongoing bleeding from the arterial puncture site may decompress into the adjacent venous
puncture site, leading to the formation of an arteriovenous fistula. This complication may be
recognized by the presence of a thrill or continuous bruit at the site of catheter insertion.
Surgical repair is usually necessary, since fistulae tend to enlarge over time. The most
common finding at surgery is a puncture site below the common femoral artery, which
illustrates the importance of careful technique in avoiding this complication [1].

3.4 Prior pseudoaneurysm


If a hematoma remains in continuity with the arterial lumen, a pseudoaneurysm may
develop with blood flow into and out of the hematoma cavity during systole and diastole. A
pseudoaneurysm is most often recognized by the presence of a pulsatile mass with a systolic
bruit over the catheter insertion site; it can be confirmed by duplex ultrasonography. Almost
all pseudoaneurysms occur within the first three days after removal of the arterial sheath,
and most of the remaining cases by seven days after sheath removal [28].
The main risk factor for pseudoaneurysm formation is an inadequate period of manual
compression. This was illustrated in a series of 581 angiographic procedures involving
transfemoral arterial catheterization [29]. A pseudoaneurysm occurred in 14 percent of the
first 300 procedures in which standard manual compression was performed, compared to
1.1 percent of the next 281 procedures when manual compression was continued for at least
five minutes after local bleeding had stopped.
Other risk factors include large bore sheaths, postprocedural anticoagulation, antiplatelet
therapy during the intervention, age >65 years, obesity, hypertension, peripheral artery
disease, hemodialysis, cannulation of the superficial rather than common femoral artery,
and complex interventions [28-30].
Surgical management should be performed when a pseudoaneurysm occurs at the site of a
vascular anastomosis, becomes very large and threatens or causes skin necrosis, is
expanding rapidly, occurs spontaneously, as it may be infected, or there is a failure of a
minimally invasive procedure [30]. Expanding hematomas have a propensity towards
eventual rupture, especially if the patient is maintained on anticoagulation [1,7,31,32].
Treatment of smaller pseudoaneurysms may be accomplished with either direct ultrasound-
guided compression or with ultrasound-guided local injection of thrombin or collagen into
the pseudoaneurysm cavity, which has a higher success rate [28,33-35]. Although an
effective approach, ultrasound-guided compression is uncomfortable for both the patient
and the person applying compression.
The success rate of ultrasound-guided injection of bovine thrombin was illustrated in a
series of 240 patients with simple or complex pseudoaneurysms [28]. The primary success
rate was 96 percent with simple pseudoaneurysms and 89 percent with complex
pseudoaneurysms. The secondary success rate was 99.6 percent (all but one patient). The
authors also reviewed 34 studies with 1388 patients; the overall success rate was 97.2
percent.
The high success rate of ultrasound-guided injection has made it the treatment of choice for
most postcatheterization pseudoaneurysms [30]. However, the procedure should not be
performed when, as defined above, there is an indication for surgical management.
In rare cases, pseudoaneurysms less than 2 cm can be managed conservatively and
monitored by serial imaging to confirm spontaneous resolution.
158 Advances in the Diagnosis of Coronary Atherosclerosis

3.5 Arterial thrombosis


Arterial thrombosis is rare and occurs slightly more often when the brachial approach is
used [3,4]. Predisposing factors to femoral artery thrombosis include small vessel lumen,
peripheral artery disease, diabetes mellitus, female sex, placement of a large diameter
catheter or sheath (eg, intraaortic balloon pump), or a long catheter dwell time [2-7,31,32].
Patients with lower extremity pain or paresthesia, plus reduced or absent distal pulses not
responding to catheter removal, should be evaluated for possible flow obstructing thrombus
or dissection. Urgent vascular surgery or thrombectomy may be required for preservation of
the limb.

3.6 Radial artery access


Local vascular complications appear to occur less frequently with radial artery access. This
issue was best addressed in a meta-analysis that evaluated 12 randomized trials including
3224 patients comparing radial with femoral access [36]. The risk of major adverse cardiac
events was similar for the two approaches. Radial access was associated with a significantly
lower incidence of access site complications (odds ratio 0.20, 95% CI 0.09-0.42), but a
significantly higher rate of procedural failure (odds ratio 3.30. 95% CI 1.63-6.71), probably
due to the specific technical skills required for this method.

3.6.1 Integrity of the palmar arch


Radial artery cannulation is associated with a 5 to 19 percent chance of radial artery
occlusion [37]. This complication is not clinically important in most patients because the
hand is perfused by both the radial and ulnar arteries with extensive collateral flow between
the two. However, some patients have incomplete palmar arches, which may diminish
collateral perfusion, possibly leading to hand ischemia in the presence of radial artery
occlusion.
The modified Allen test has traditionally been used to identify patients at increased risk
from radial artery catheterization. However, many centers now use pulse oximetry and
plethysmography.
The modified Allen test, originally developed to evaluate patients with thromboangiitis
obliterans, assesses the adequacy of collateral blood supply from the ulnar artery to the
hand via the palmar arches. The test consists of the following steps:
 The patient's hand is initially held high while the fist is clenched and both radial and
ulnar arteries are compressed; this allows the blood to drain from the hand.
 The hand is then lowered and the fist is opened.
 After pressure is released over the ulnar artery, color should return to the hand within
six seconds, indicating a patent ulnar artery and an intact superficial palmar arch. The
test is considered abnormal if 10 seconds or more is required for color to return.
In two large series of patients undergoing cardiac catheterization, an abnormal Allen test
result was obtained in 6 and 27 percent [38,39]. The importance of an abnormal test was
evaluated in a study of 55 patients undergoing coronary angiography, 20 of whom had an
abnormal test [37]. After 30 minutes of subsequent radial artery occlusion, the patients with
an abnormal test had significantly reduced blood flow to the thumb and an elevated thumb
capillary lactate.
Based upon such observations, radial artery catheterization is usually not performed in
patients with an abnormal modified Allen test. A possible exception occurs in patients with
Complications of Cardiac Catetherization 159

an excessive estimated risk from a femoral approach (eg, severe peripheral artery disease,
large abdominal aortic aneurysm, morbid obesity) [37].
The results of the modified Allen test can be altered by a number of factors, including
overextension of the wrist, skin tension over the ulnar artery, and operator error. As a result,
an appreciable number of false positive and false negative results have been reported [38].
As an alternative, many centers use a more direct assessment of blood flow to the thumb
during radial artery occlusion, which can be accomplished with pulse oximetry and
plethysmography [38,40].
The traditional modified Allen test was compared to pulse oximetry and plethysmography
in a consecutive series of 1010 patients referred for diagnostic cardiac catheterization [38].
The modified Allen test was considered abnormal if palmar blanching persisted for ≥10
seconds after release of ulnar compression. Plethysmography was observed for two minutes
during radial artery compression. Plethysmography was characterized as follows:
 A: no change in the amplitude of the pulse tracing during compression
 B: reduction in amplitude with compression
 C: loss of pulse tracing with initial compression, but recovery of flow during two
minutes of compression (signifying development of collateral flow)
 D: loss of pulse tracing with no recovery
Patients in categories A, B and C were considered to have a patent ulnopalmar arch, and
therefore eligible for radial catheterization.
The modified Allen test was abnormal in significantly more patients than plethysmography
(6.3 versus 1.5 percent). The authors concluded that plethysmography more accurately
demonstrates the adequacy of the ulnopalmar arch, and that the modified Allen test may
unnecessarily exclude some patients from radial artery catheterization. However, the
number of patients who underwent radial artery catheterization was not reported, and the
value of either test for predicting ischemic complications following radial artery
catheterization was not assessed.

4. Arrhythmias
A variety of arrhythmias and conduction disturbances can occur during catheterization. The
most common, ventricular premature beats, may be induced by catheter introduction into
the right or left ventricle, and are generally without clinical importance or impact.

4.1 Ventricular tachycardia or fibrillation


Ventricular tachycardia or fibrillation is rare complications of catheterization, occurring in
0.4 percent of cases in the second registry of the Society for Cardiac Angiography [2]. These
arrhythmias may result from excess catheter manipulation or, more commonly, from
intracoronary contrast injection. This complication occurs more commonly when ionic and
hyperosmolar contrast agents are used.

4.2 Atrial arrhythmias


Atrial arrhythmias can be induced by cardiac catheterization, especially in response to
catheter placement into or out of the right atrium during right heart catheterization. These
arrhythmias usually do not require immediate treatment unless they produce ischemia or
hemodynamic instability.
160 Advances in the Diagnosis of Coronary Atherosclerosis

Atrial flutter is generally well tolerated and can be treated with burst atrial pacing, or
electrical or pharmacologic cardioversion. Atrial fibrillation, on the other hand, can cause
clinical sequelae in the setting of a rapid ventricular response or if the loss of atrial systole
results in hypotension, as in a patient with mitral stenosis or diastolic ventricular
dysfunction. Synchronized cardioversion should be performed immediately in patients who
are hemodynamically unstable.

4.3 Bradycardia
Bradycardia commonly occurs during coronary angiography, most often at the end of the
injection of ionic contrast into the right coronary artery. Forceful coughing helps to clear the
contrast, support perfusion, and restore normal cardiac rhythmtachycardia or fibrillation is
rare complications of catheterization, occurring in 0.4 percent of cases in the second registry of
the Society for Cardiac Angiography [2]. These arrhythmias may result from excess catheter
manipulation or, more commonly, from intracoronary contrast injection. This complication
occurs more commonly when ionic and hyperosmolar contrast agents are used.
Vasovagal reactions, which can include bradycardia, hypotension, yawning, and/or sweating,
are seen in up to 3 percent of patients, particularly if they have pain or are anxious [2]. These
reactions may occur with palpation or compression of the artery prior to needle access.
Treatment usually consists of volume administration, atropine (0.5 to 1.0 mg intravenously),
and removal of the painful stimulus. Patients with critical coronary or valvular disease may
undergo irreversible decompensation from vagally-mediated hypotension. As a result, rapid
reversal of hypotension is crucial in this setting.

4.4 Conduction disturbances


Conduction disturbances are a rare cause of bradycardia in the catheterization laboratory.
Patients with pre-existing left bundle branch block may develop asystole with right heart
catheterization. Patients with pre-existing right bundle branch block may develop asystole
during left heart catheterization. Atropine is rarely helpful in patients who develop
complete heart block, but its administration is still recommended. Coughing may help
support the circulation while the right heart catheter is exchanged for a temporary pacing
wire. Prophylactic placement of a temporary pacemaker is not necessary, even in patients
with bundle branch block or intervention in the right coronary artery.
Frank asystole is rare and there is generally sufficient time for insertion of a pacing wire
through a venous sheath [41].

5. Perforation of the heart or great vessels


Perforation is an extremely rare complication of cardiac catheterization. Higher risk
procedures are those that involve stiffer catheters, including transseptal catheterization,
endomyocardial biopsy, balloon valvuloplasty, needle pericardiocentesis, and placement of
a pacing catheter. Older adult women appear to be at particular risk.
Cardiac perforation is generally heralded by bradycardia and hypotension due to vagal
stimulation induced by blood in the pericardium. If the patient is hemodynamically stable,
echocardiography is suggested to document the presence of blood in the pericardial space.
However, emergent pericardiocentesis should be performed via the subxiphoid approach in
the presence of hemodynamic instability.
Complications of Cardiac Catetherization 161

Since pericardiocentesis is itself associated with complications, it is best performed under


echocardiographic guidance or with pressure monitoring. In a study of 91 cases of acute
tamponade during catheterization, pericardiocentesis was the only therapy necessary in 82
percent [42].
Reversal of anticoagulation may cause blood in the pericardial space to "gel." As a result, we
generally try to aspirate the majority of the blood in the pericardial space prior to the
administration of protamine [1].

6. Allergic reactions
Allergic reactions during catheterization may be precipitated by three agents: the local
anesthetic, the contrast agent, or protamine sulfate.

6.1 Local anesthetic


In patients claiming previous reaction to a local anesthetic, the use of a preservative-free
agent (such as bupivacaine or mepivacaine) may be a safe alternative [1].

6.2 Iodinated contrast agents


Reaction to the contrast agent occurs in up to 1 percent of patients [2-7]. The risk of such a
reaction is highest in patients with a history of prior contrast reactions. Other conditions,
such as asthma and other atopic diseases, also confer some risk.
There are several steps that can reduce the incidence of a reaction in a high-risk patient:
 Premedication with corticosteroids and H1 antihistamines can reduce the incidence of a
severe reaction (eg, bronchospasm or shock) to below 1 percent (table 4).
 The patient should receive a nonionic contrast agent. The incidence of crossreactivity in
patients who have had previous reactions to older agents is low, although all patients
with previous reactions should be premedicated [1]. In the past, nonionic contrast
agents were used selectively, given their higher cost.
Prednisone 50 mg orally 13,7, and 1 h prior to procedure
[In children: 0.5 to 0.7 mg/kg orally, up to 50 mg]
Diphenhydramine 50 mg PO/IM/ or IV 1 h prior to procedure
[In children: 1.25 mg/kg orally, up to 50 mg]
Lower / iso-osmolar RCM should be recommended
Emergency therapy should be available
Table 4. Pretreatment for patients with previous adverse reactions to RCM
However, the cost of nonionic contrast has fallen significantly. Thus, we recommend using
nonionic contrast in all patients undergoing coronary angiography.
If a severe reaction does occur, it should be treated in the same manner as anaphylaxis, and
can be sometimes be reversed by using boluses of intravenous epinephrine (1:10,000
epinephrine so that 1 mL = 0.1 mg). One mL of this solution should be administered
approximately every minute until the arterial pressure is restored.
However, the rapidity of dosing must be adjusted by the degree of hypotension and the
level of assessed hypoperfusion. It is rare for a patient to require a total dose of more than
1.0 mg (10 mL).
Contrast-induced acute renal failure represents another complication that is discussed
below.
162 Advances in the Diagnosis of Coronary Atherosclerosis

6.3 Protamine
Allergic reactions to protamine sulfate occur occasionally, more often in insulin-dependent
diabetic patients who have received NPH insulin (which contains protamine) in the past [1].
The rapid administration of protamine sulfate can also provoke back pain of unknown
etiology.
In current practice, protamine is rarely administered. Most diagnostic catheterizations are
done without heparin anticoagulation and in most interventional procedures the heparin is
either allowed to wear off over the next two to three hours prior to sheath removal, or the
sheath is removed in the fully anticoagulated state using an arterial closure device.

7. Atheroembolism
During the process of cardiac catheterization, atheromatous debris is often scraped from the
aortic wall. This debris can cause systemic embolization, including cutaneous, renal, retinal,
cerebral, and gastrointestinal emboli (photo 1 and photo 2 and photo 3). To minimize the
risk of stroke or visual impairment, arterial catheter exchanges are best performed over a
wire in the descending aorta at the level of the diaphragm (for closed loop catheters) and in
the ascending aorta for angiographic catheters. Once arterial access has been obtained, the
practice of readvancing the guidewire through the infrarenal aorta rather than exchanging it
through a catheter should be avoided.

Blue toes are a classic manifestation of peripheral embolization of atheromatous material from proximal
arterial sources (eg, aorta); the pedal pulses are often normal. This patient, who has a 30-year history of
type 1 diabetes and severe peripheral vascular disease, presented with foot pain and discoloration.
Cholesterol microemboli from the aorta were suspected to be the cause.
Photo 1. Blue toe syndrome
Complications of Cardiac Catetherization 163

The high rate of release of atheromatous debris was illustrated in a series of 1000 patients
undergoing percutaneous coronary intervention: 51 percent had aortic atheromatous
material retrieved from the catheter after back flow of blood [43]. There were no associated
in-hospital ischemic complications, probably due to sufficient withdrawal of blood
containing the debris prior to the injection of contrast.
The reported incidence of atheroembolic events varies from 0.6 to 0.9 percent in
retrospective studies to 1.4 to 1.9 percent in prospective series [44].

Patient with lupus and anti-phospholipid antibodies with livedo reticularis (manifested by a reddish-
cyanotic, reticular pattern of the skin) which has resulted in ulcer formation (arrows).
Photo 2. Livedo reticularis
The clinical features of atheroembolism after diagnostic cardiac catheterization were
examined in a prospective evaluation of 1786 patients ≥40 years of age [45]. The following
observations were made:
 25 patients (1.4 percent) were diagnosed with atheroembolism; 12 had cutaneous signs
(eg, "blue toes" syndrome, livedo reticularis) (photo 1 and photo 2) and 16 had acute
renal failure that was considered to be atheroembolic in origin because it persisted for
at least two weeks (while contrast nephropathy typically reverses within the first week).
 Eosinophil counts were significantly higher in patients suffering from atheroembolism
both before and after catheterization.
 Four patients with atheroembolism died in hospital (16 percent); all had progressive
renal dysfunction. The mortality rate was much lower (0.5 percent) in the patients
without atheroembolism.
164 Advances in the Diagnosis of Coronary Atherosclerosis

Photo 3. Hollenhorst plaque (cholesterol crystal, arrow) in retinal artery

8. Acute renal failure


There are three major causes of renal dysfunction related to cardiac catheterization: contrast-
induced acute renal failure, renal atheroemboli, and hemodynamic instability with renal
hypoperfusion.

8.1 Contrast nephropathy


At least 5 percent of patients who undergo cardiac catheterization experience a transient rise
in the plasma creatinine concentration of more than 1.0 mg/dL (88 micromol/L) due to
contrast-induced renal dysfunction [46].
However, there is substantial variability in risk among patients, some of whom have a risk
that exceeds 50 percent. The risk is greatest in patients with moderate to severe renal
insufficiency and diabetes.
The plasma creatinine concentration usually returns to baseline within seven days, and less
than 1 percent of patients, usually diabetic patients with underlying severe chronic kidney
disease, go on to require chronic hemodialysis.
Nonionic low osmolal contrast agents are now administered for the majority of radiologic
procedures using intravascular contrast media, as a result of increased patient tolerability and
decreased hypersensitivity reactions. However, in patients at risk for contrast nephropathy, we
Complications of Cardiac Catetherization 165

prefer the use of iso-osmolal agents (approximately 290 mosmol/kg) rather than low osmolal
agents (500 to 850 mosmol/kg) because of a lower risk of contrast nephropathy.
The 2007 focused update of the ACC/AHA/SCAI 2005 PCI guidelines recommended the use
of iso-osmolal in preference to low osmolal agents in patients with chronic kidney disease [47].

8.2 Renal atheroemboli


The kidney is one of the principal end organs that may be affected by atheroembolic events.
(See 'Atheroembolism' above.) There are three main findings that suggest that atheroemboli
rather than contrast nephropathy is responsible for the development of acute renal failure
after catheterization:
 The presence of other signs of embolization such as blue toes (photo 1), livedo reticularis
(photo 2), Hollenhorst plaques in the retina (photo 3), and/or abdominal pain.
 Transient eosinophilia and hypocomplementemia.
 Persistent renal failure after seven days.
In a review of 1786 consecutive patients undergoing cardiac catheterization, acute renal
failure developed in 0.9 percent overall and in 64 percent of those with evidence of any
atheroembolism [45]. In this report, acute renal failure was considered to be due to renal
atheroembolism if it had persisted for at least two weeks, well beyond the time at which
contrast nephropathy should have resolved.

9. Infection
Cardiac catheterization procedures are performed using sterile technique. Thus, both local
infection and bacteremia are rare and endocarditis prophylaxis during cardiac
catheterization is not recommended for patients with valvular disease. Infection is
approximately 10 times more common with the brachial approach compared to the femoral
approach (0.6 versus 0.06 percent) [2-7].
Infection control guidelines for the cardiac catheterization laboratory were updated in 2006
by the Society for Cardiovascular Angiography and Interventions [48]. These guidelines
emphasize the importance of handwashing caps, gowns, gloves, and masks to protect the
patient against bacterial infection, and measures such as eye wear and vaccination for
hepatitis B to protect laboratory personnel.

10. Radiation exposure


Patients undergoing diagnostic and interventional cardiac catheterization receive substantial
radiation exposure.

11. Endomyocardial biopsy


Endomyocardial biopsy of right ventricular tissue is made using a relatively stiff catheter.
Complications such as perforation of the heart or great vessels, life threatening arrhythmias,
or heart block requiring permanent pacing occur uncommonly.

12. Conclusions
Cardiac catheterization is a common proceduce with very low incidence of complications
nowadays. We can consider different kind of adverse events. On the one hand, the events
166 Advances in the Diagnosis of Coronary Atherosclerosis

related with the artery access, that are the most common, like haematoma, pseudoaneurysm
or dissection. On the other hand, we can observe complications more severe like
embolizations, renal failure or allergic reactions. Death is an uncommon complication, with
a is generally well below 1 percent. It is very important to know the potential adverse
events of these procedures in order to be aware to prevent it as better as possible.

13. References
[1] Baim, DS, Grossman, W. Complications of cardiac catheterization. In: Cardiac
Catheterization, Angiography and Intervention, Baim, DS, Grossman, W (Eds),
Williams & Wilkins, Baltimore 1996. p.17.
[2] Johnson LW, Lozner EC, Johnson S, et al. Coronary arteriography 1984-1987: a report of
the Registry of the Society for Cardiac Angiography and Interventions. I. Results
and complications. Cathet Cardiovasc Diagn 1989; 17:5.
[3] Dorros G, Cowley MJ, Simpson J, et al. Percutaneous transluminal coronary angioplasty:
report of complications from the National Heart, Lung, and Blood Institute PTCA
Registry. Circulation 1983; 67:723.
[4] Kennedy JW. Complications associated with cardiac catheterization and angiography.
Cathet Cardiovasc Diagn 1982; 8:5.
[5] Noto TJ Jr, Johnson LW, Krone R, et al. Cardiac catheterization 1990: a report of the
Registry of the Society for Cardiac Angiography and Interventions (SCA&I). Cathet
Cardiovasc Diagn 1991; 24:75.
[6] Laskey W, Boyle J, Johnson LW. Multivariable model for prediction of risk of significant
complication during diagnostic cardiac catheterization. The Registry Committee of
the Society for Cardiac Angiography & Interventions. Cathet Cardiovasc Diagn
1993; 30:185.
[7] Wyman RM, Safian RD, Portway V, et al. Current complications of diagnostic and
therapeutic cardiac catheterization. J Am Coll Cardiol 1988; 12:1400.
[8] King KM, Ghali WA, Faris PD, et al. Sex differences in outcomes after cardiac
catheterization: effect modification by treatment strategy and time. JAMA 2004;
291:1220.
[9] Hemmelgarn BR, Southern DA, Humphries KH, et al. Refined characterization of the
association between kidney function and mortality in patients undergoing cardiac
catheterization. Eur Heart J 2006; 27:1191.
[10] Gruberg L, Mintz GS, Mehran R, et al. The prognostic implications of further renal
function deterioration within 48 h of interventional coronary procedures in patients
with pre-existent chronic renal insufficiency. J Am Coll Cardiol 2000; 36:1542.
[11] Segal AZ, Abernethy WB, Palacios IF, et al. Stroke as a complication of cardiac
catheterization: risk factors and clinical features. Neurology 2001; 56:975.
[12] Hamon M, Baron JC, Viader F, Hamon M. Periprocedural stroke and cardiac
catheterization. Circulation 2008; 118:678.
[13] Omran H, Schmidt H, Hackenbroch M, et al. Silent and apparent cerebral embolism
after retrograde catheterisation of the aortic valve in valvular stenosis: a
prospective, randomised study. Lancet 2003; 361:1241.
[14] Chambers J, Bach D, Dumesnil J, et al. Crossing the aortic valve in severe aortic stenosis:
no longer acceptable? J Heart Valve Dis 2004; 13:344.
Complications of Cardiac Catetherization 167

[15] Büsing KA, Schulte-Sasse C, Flüchter S, et al. Cerebral infarction: incidence and risk
factors after diagnostic and interventional cardiac catheterization--prospective
evaluation at diffusion-weighted MR imaging. Radiology 2005; 235:177.
[16] Lund C, Nes RB, Ugelstad TP, et al. Cerebral emboli during left heart catheterization
may cause acute brain injury. Eur Heart J 2005; 26:1269.
[17] Pracyk JB, Wall TC, Longabaugh JP, et al. A randomized trial of vascular hemostasis
techniques to reduce femoral vascular complications after coronary intervention.
Am J Cardiol 1998; 81:970.
[18] Ward SR, Casale P, Raymond R, et al. Efficacy and safety of a hemostatic puncture
closure device with early ambulation after coronary angiography. Angio-Seal
Investigators. Am J Cardiol 1998; 81:569.
[19] Kapadia SR, Raymond R, Knopf W, et al. The 6Fr Angio-Seal arterial closure device:
results from a multimember prospective registry. Am J Cardiol 2001; 87:789.
[20] Silber S, Gershony G, Schön B, et al. A novel vascular sealing device for closure of
percutaneous arterial access sites. Am J Cardiol 1999; 83:1248.
[21] Silber S, Tofte AJ, Kjellevand TO, et al. Final report of the European multi-center
registry using the Duett vascular sealing device. Herz 1999; 24:620.
[22] Baim DS, Knopf WD, Hinohara T, et al. Suture-mediated closure of the femoral access
site after cardiac catheterization: results of the suture to ambulate aNd discharge
(STAND I and STAND II) trials. Am J Cardiol 2000; 85:864.
[23] Koreny M, Riedmüller E, Nikfardjam M, et al. Arterial puncture closing devices
compared with standard manual compression after cardiac catheterization:
systematic review and meta-analysis. JAMA 2004; 291:350.
[24] Nikolsky E, Mehran R, Halkin A, et al. Vascular complications associated with
arteriotomy closure devices in patients undergoing percutaneous coronary
procedures: a meta-analysis. J Am Coll Cardiol 2004; 44:1200.
[25] Geary K, Landers JT, Fiore W, Riggs P. Management of infected femoral closure
devices. Cardiovasc Surg 2002; 10:161.
[26] Cherr GS, Travis JA, Ligush J Jr, et al. Infection is an unusual but serious complication
of a femoral artery catheterization site closure device. Ann Vasc Surg 2001; 15:567.
[27] El-Jack SS, Ruygrok PN, Webster MW, et al. Effectiveness of manual pressure
hemostasis following transfemoral coronary angiography in patients on
therapeutic warfarin anticoagulation. Am J Cardiol 2006; 97:485.
[28] Krueger K, Zaehringer M, Strohe D, et al. Postcatheterization pseudoaneurysm: results
of US-guided percutaneous thrombin injection in 240 patients. Radiology 2005;
236:1104.
[29] Katzenschlager R, Ugurluoglu A, Ahmadi A, et al. Incidence of pseudoaneurysm after
diagnostic and therapeutic angiography. Radiology 1995; 195:463.
[30] Webber GW, Jang J, Gustavson S, Olin JW. Contemporary management of
postcatheterization pseudoaneurysms. Circulation 2007; 115:2666.
[31] Muller DW, Shamir KJ, Ellis SG, Topol EJ. Peripheral vascular complications after
conventional and complex percutaneous coronary interventional procedures. Am J
Cardiol 1992; 69:63.
[32] Popma JJ, Satler LF, Pichard AD, et al. Vascular complications after balloon and new
device angioplasty. Circulation 1993; 88:1569.
168 Advances in the Diagnosis of Coronary Atherosclerosis

[33] Paulson EK, Sheafor DH, Kliewer MA, et al. Treatment of iatrogenic femoral arterial
pseudoaneurysms: comparison of US-guided thrombin injection with compression
repair. Radiology 2000; 215:403.
[34] La Perna L, Olin JW, Goines D, et al. Ultrasound-guided thrombin injection for the
treatment of postcatheterization pseudoaneurysms. Circulation 2000; 102:2391.
[35] Hamraoui K, Ernst SM, van Dessel PF, et al. Efficacy and safety of percutaneous
treatment of iatrogenic femoral artery pseudoaneurysm by biodegradable collagen
injection. J Am Coll Cardiol 2002; 39:1297.
[36] Agostoni P, Biondi-Zoccai GG, de Benedictis ML, et al. Radial versus femoral approach
for percutaneous coronary diagnostic and interventional procedures; Systematic
overview and meta-analysis of randomized trials. J Am Coll Cardiol 2004; 44:349.
[37] Greenwood MJ, Della-Siega AJ, Fretz EB, et al. Vascular communications of the hand in
patients being considered for transradial coronary angiography: is the Allen's test
accurate? J Am Coll Cardiol 2005; 46:2013.
[38] Barbeau GR, Arsenault F, Dugas L, et al. Evaluation of the ulnopalmar arterial arches
with pulse oximetry and plethysmography: comparison with the Allen's test in
1010 patients. Am Heart J 2004; 147:489.
[39] Benit E, Vranckx P, Jaspers L, et al. Frequency of a positive modified Allen's test in 1,000
consecutive patients undergoing cardiac catheterization. Cathet Cardiovasc Diagn
1996; 38:352.
[40] Hovagim AR, Katz RI, Poppers PJ. Pulse oximetry for evaluation of radial and ulnar
arterial blood flow. J Cardiothorac Anesth 1989; 3:27.
[41] Harvey JR, Wyman RM, McKay RG, Baim DS. Use of balloon flotation pacing catheters
for prophylactic temporary pacing during diagnostic and therapeutic
catheterization procedures. Am J Cardiol 1988; 62:941.
[42] Tsang TS, Freeman WK, Barnes ME, et al. Rescue echocardiographically guided
pericardiocentesis for cardiac perforation complicating catheter-based procedures.
The Mayo Clinic experience. J Am Coll Cardiol 1998; 32:1345.
[43] Keeley EC, Grines CL. Scraping of aortic debris by coronary guiding catheters: a
prospective evaluation of 1,000 cases. J Am Coll Cardiol 1998; 32:1861.
[44] Bashore TM, Gehrig T. Cholesterol emboli after invasive cardiac procedures. J Am Coll
Cardiol 2003; 42:217.
[45] Fukumoto Y, Tsutsui H, Tsuchihashi M, et al. The incidence and risk factors of
cholesterol embolization syndrome, a complication of cardiac catheterization: a
prospective study. J Am Coll Cardiol 2003; 42:211.
[46] Tommaso CL. Contrast-induced nephrotoxicity in patients undergoing cardiac
catheterization. Cathet Cardiovasc Diagn 1994; 31:316.
[47] King SB, 3rd, Smith, SC Jr, Hirshfeld, JW Jr, et al. 2007 Focused Update of the
ACC/AHA/SCAI 2005 Guideline Update for Percutaneous Coronary Intervention:
a report of the American College of Cardiology/American Heart Association Task
Force on Practice Guidelines: 2007 Writing Group to Review New Evidence and
Update the ACC/AHA/SCAI 2005 Guideline Update for Percutaneous Coronary
Intervention, Writing on Behalf of the 2005 Writing Committee. Circulation 2008;
117:261.
[48] Chambers CE, Eisenhauer MD, McNicol LB, et al. Infection control guidelines for the
cardiac catheterization laboratory: society guidelines revisited. Catheter Cardiovasc
Interv 2006; 67:78.
9

Diagnosis and Management of


Complications of Invasive
Coronary Angiography
Jong-Seon Park and Young-Jo Kim
Yeungnam University Hospital
Korea

1. Introduction
Since its introduction several decades ago, coronary angiography and intervention has
become a widely and ever increasingly popular diagnostic and therapeutic modality.
However, it is not devoid of occasional systemic and local complications, and their
immediate detection and management are crucial to achieving a favourable clinical course.
This chapter focuses on the mechanism, diagnosis and treatment of the inherent risks of
coronary angiography and intervention.

2. Systemic complications
Coronary angiography and intervention comprise local procedures performed via a
percutaneous vascular approach. Untoward hemodynamic and systemic hormonal changes
and toxic effects of the contrast agents used might occur during and after procedures.
Patient’s general status, comorbid conditions and degree of systemic atherosclerosis are the
most important factors predisposing to the development of systemic complications.
Effective pre-procedural preparation and immediate proper treatment help prevent event
occurrence and its subsequent deterioration to permanent sequelae.

2.1 Side effects of contrast agent


Use of contrast agents has a long history since the introduction of X-ray technology in 1895,
and progressive changes after Douglas Cameron of Minnesota recommended the use of
sodium iodide for retrograde phelography reduced toxicity leading to current angiography
practices. The side effects of contrast agents are related to their osmolarity, hydrophilicity
and viscosity, and there currently are different kinds of contrast media available which can
be classified according to their chemical characteristics (Table 1).
Osmolarity is the most important characteristic of contrast agents, which thereby are
classified into high-osmolar (∼2,000 mOsm/kg), low-osmolar (600∼800 mOsm/kg) and iso-
osmolar (290 mOsm/kg) contrast media. Commonly, high osmolar contrast agents induce
fluid shift into vessels, which can cause a febrile sensation and pain. Also, high osmolar
contrast media take cellular fluid from cells including red blood cells and therefore lead to
dehydration and morphological changes. Hemodilution and hypervolemia can further
170 Advances in the Diagnosis of Coronary Atherosclerosis

aggravate heart failure when present. In the past 60 years, high–osmolar contrast media
with osmolarity 5∼8 times higher than that of plasma have been replaced by low-osmolar
contrast media having osmolarity 2∼3 times higher than that of plasma.

Osmolarity Molecular Component Iodine Osmolarity Viscosity


structure concentration (mOsmol/kg) at 37oC
(mg/mL) (cps)
High- Ionic Sodium 320-370 1,500∼2,100 2.75-5.0
osmolar monomer iothalamate
Meglumine
diatrizoate
Low- Ionic Meglumine 320 580 6
osmolar dimer ioxaglate
Iopamidol 300 520-672 4.5-6.3
Iohexol
Loversol
Iopromide
Iso- Nonionic Iotrolan 240 320 8.1
osmolar dimer Iodixanol 320 290 11.4
Table 1. Characteristics of contrast media
Cell membranes consist of phospholipid bilayers which are resistant to hydrophilic
molecules. Therefore hydrophilic contrast agents are safer than hydrophobic ones.
Viscosity increases as iodine concentration increases and temperature decreases; therefore,
contrast agents should be heated from room temperature to body temperature before use.
Early ionic contrast agents were associated with high incidence of early systemic side effects
such as nausea, urticaria, pain, and heat sensation by comparison to non-ionic agents.
Therefore, ionic agents have been replaced with non-ionic agents. However, the incidence of
late skin reaction, i.e., 24 hours after injection of non-ionic dimeric agents is relatively high
as compared to monomeric agents (Sutton et al 2001).
The side effects of contrast media are divided into anaphylactoid and non-anaphylactoid
reactions. Most of the adverse effects occur within 20 minutes after administration of
contrast agents. Therefore, the patient’s systemic symptoms, blood pressure, heart rate and
rhythm, and respiratory status should be carefully monitored early after contrast injection.
Furthermore, the catheterization laboratory always should be ready for cardiopulmonary
resuscitation.

2.1.1 Anaphylactoid reactions


Anaphylactoid reaction is a life-threatening side-effect of contrast media which occurs
within 20 minutes and requires immediate, intensive therapy; the incidence of use of high
osmolar contrast agent is approximately 1∼3 % and a fatal reaction has been estimated at 0.9
cases per 100,000 exposures (Krause 2010). The term anaphylactoid reaction refers to a
syndrome that albeit clinically similar to anaphylaxis differs from true hypersensitivity
reactions by mast cell or basophil degranulation which is independent from
immunoglobulin-E mediation, not requiring previous sensitisation, and not consistently
recurring in an experienced patients (Hong et al 2002). Mild symptoms include urticaria,
Diagnosis and Management of Complications of Invasive Coronary Angiography 171

pruritus, dizziness, mild dyspnea, nausea and vomiting and these are usually self-limiting
and resolving with minimal medical therapy. In severe cases, symptoms such as
hypotension, bronchospasm, laryngeal edema, abdominal cramps, pulmonary edema,
syncope, and serious arrhythmias can occur and lead to death and therefore require
intensive treatment. In patients who are at high risk of an anaphylactoid reaction to contrast
media, H1 antihistamines and corticosteroids prior to the administration of ionic contrast
may be useful in preventing anaphylactoid reactions to contrast agents (Delaney et al 2006).
The treatment of anaphylactoid reactions depends on the severity of symptoms and clinical
findings. Monitoring of the respiratory status and oxygen concentration is very important. If
mild bronchospasm occurs, patient can be treated with oxygen supplementation and/or
bronchodilator inhalation. In moderate cases, 0.1-0.3 mL of 1:1000 epinephrine should be
subcutaneously administered carefully, especially in patients with cardiac disease. In more
severe cases, 1 mL of 1:10,000 epinephrine can be administered intravenously for 5 minutes,
which can be repeated every 10 minutes. Additionally, antihistamines may be administered.
Hypotension is another serious side effect of contrast agents. Immediate oxygen
supplementation and isotonic fluid supplement is necessary, and if ineffective, dopamine at
an infusion rate of 2-20 μgm/kg/min can be administered.

2.1.2 Non-anaphylactoid reactions


Early (<24 hours) and late (24 hours ∼ 7 days) reactions may occur after injection of contrast
agents. The mechanisms of the late reactions are unknown and their manifestations are very
diverse and non-specific, including flulike symptoms such as general weakness, headache,
fever, chill, vomiting, abdominal pain, constipation or diarrhea, pruritus and rash. Most
symptoms are self-limiting and respond well to symptomatic treatment and fluid
administration.
Hypotension and bradycardia as consequence of vasovagal stimulation associated with
high-osmolar contrast infusion are common. Isotonic fluid and atropine administration
(0.5∼1 mg, intravenously, repeated every 3-5 minutes) are effective in most cases. Life
threatening cardiac arrhythmias such as ventricular tachycardia and ventricular fibrillation
can occur after rapid injection of contrast media, especially in patients with acute
myocardial infarction. Careful ECG monitoring after first contrast injection and immediate
cardioversion or defibrillation should be performed.
Contrast-induced nephropathy (CIN), an important complication in the use of contrast
media, is associated with increased frequency of late cardiovascular events after coronary
intervention as well as increased risk of death. The commonly used definition of CIN is a
rise in serum creatinine level of 0.5 mg/dL or a 25% increase from baseline value, assessed
at 48 hours after angiography (Solomon et al 2010). The incidence of CIN in patients
undergoing coronary intervention varies depending on the definition, preventive
management, baseline renal function and procedures, and it ranges between 2∼37%
(Solomon et al. 2010, Gruberg et al 2000). There are several known risk factors for CIN
(Table 2). The type of contrast media used is one of the important factors for the
development or aggravation of renal failure. A meta-analysis showed that low-osmolarity
contrast media was associated with significantly lower rates of CIN than high-osmolarity
contrast media, especially in patients with pre-existing renal failure (Barrett et al 1993).
Also, iso-osmolar contrast media have been shown to be associated with a lower risk of CIN
than low-osmolar contrast media in patients with high risk for the development of CIN.
172 Advances in the Diagnosis of Coronary Atherosclerosis

Patient comorbidities Diabetes mellitus


Congestive heart failure
Acute hypotension
Anemia
Myocardial infarction
Nephrotoxic drugs
Volume depletion
Reduced renal function (eGFR <60 mL/min/1.73 m)
Procedural factors Contrast volume
Type of contrast
Periprocedural hypotension
Table 2. Risk factors for contrast-induced nephropathy(Solomon et al. 2010, McCullough
2008)
The pathophysiology of CIN remains unknown; however, it is believed to result from
release of acute vasoconstrictors induced by adenosine, endothelin, prostaglandin leading to
ischemic injury and death of renal tubular cells (McCullough 2008). Although more clinical
studies are needed, some effective preventive measures have been suggested based on
postulated mechanisms and clinical trial results (Table 3). Volume expansion is the easiest
and most effective method although there are limited data on optimal solution and volume.
Although many pharmacological agents including antioxidants, ascorbic acid, statins,
prostaglandins, aminophylline/theophyllines and N-acetylcysteine (NAC) have been
suggested for the prevention of CIN, none has been proven effective. Also, there are
inconsistent data regarding the renal protective effect of hydration, N-acetylcysteine, and
theophylline. Among them, NAC is considered as a cost effective and safe agent, and the
combination of volume expansion and NAC is more effective than NAC alone (Briguori et al
2007). A meta-analysis showed N-acetylcysteine as more renoprotective than hydration
alone; theophylline also might reduce risk for contrast-induced nephropathy however the
detected association did not achieve statistical significance (Kelly et al 2008).

Pre- and post-procedure Hold nephrotoxic agents


- Anti-inflammatory drugs
- High dose diuretics
- Aminoglycosides
Volume expansion (Isotonic saline with/without
bicarbonate solution)
- 1.0 mL /kg/hr, 12 hours before and 12 hours
after procedure
- Urine flow : >150 mL/hr
N-acetylcysteine
- 1200 mg oral bid pre-and post-procedure
During procedure Use iso- or low-osmolar contrast
Lowest dose of contrast
Post-procedure Dialysis and hemofiltration
Table 3. Suggestive preventive measures for contrast-induced nephropathy in patients at
risk
Diagnosis and Management of Complications of Invasive Coronary Angiography 173

2.2 Cholesterol embolization syndrome


Cholesterol embolization syndrome (CES), known as blue toe syndrome, is caused by
systemic embolization of cholesterol crystals from aortic atherosclerotic plaques. CES is
suggested by the gradual onset of peripheral cutaneous manifestations, livedo reticularis,
accompanied by progressive increases in blood urea nitrogen and creatinine levels following
an arterial catheterization procedure. The aortic plaques easily come off the aortic wall and
embolize to the brain, eyes, kidneys and extremities. Eosinophilia which is present in up to
80% of cases might be an important clue to early detection of cholesterol embolization
syndrome (Wilson et al 1991). The clinical incidence is 0.09∼1.4%, varying according to the
aggressiveness of procedures such as diagnostic angiography, coronary intervention and
peripheral vascular intervention (Fukumoto et al 2003, Scolari et al 1996). Elevation of
baseline plasma C-reactive protein (CRP) level is a risk factor independently associated with
CES indicating an important association between systemic inflammation and CES
(Fukumoto et al. 2003). The clinical presentation varies from asymptomatic to multi-organ
failure. In-hospital mortality is 16% which is increased to 70∼90% in patients with diffuse
embolism and multi-organ failure (Fine et al 1987). When CES is suspected, it is
recommended to discontinue anticoagulation because several case reports have suggested
that patients are more likely to develop CES when they are anticoagulated (Bruns et al 1978).
Most of the treatments are supportive with symptomatic measures.

2.3 Stroke
The cerebral embolism rate associated with coronary angiography is dependent on the
clinical characteristics of the patients, catheterization methods, imaging modalities for the
diagnosis, and clinical severity. In one series, silent ischemic cerebral lesions detected by
diffusion-weighted MRI were found in 5-22% patients after cardiac catheterization (Hamon
et al 2006, Busing et al 2005). After diagnostic coronary angiography via femoral approach,
stroke occurred in 0.11%, with a persistent neurological defect in only 0.04% (Ammann et al
2003). Although radial access for coronary angiography is more prone than femoral access to
generating more particulate cerebral microemboli which can pass the right middle cerebral
artery, femoral and radial access are associated with similar rates of stroke.

3. Local complications
Local vascular puncture and closure necessary in all catheterization procedures can cause a
variety of local complications, ranging from minor problems resolving with time to major
complications with long-term sequelae. For the individual patient, the risk is dependent
upon cardiovascular anatomy, the experience of the operator, and the type of procedure
being performed. Immediate detection and proper treatment is vital for the prevention of
catastrophic events.

3.1 Local hematoma and bleeding


Local bleeding and hematoma at the arterial puncture site are the most common
complications of angiography. Small hematomas at the femoral puncture site are relatively
common and usually insignificant; however, severe bleeding and hematoma requiring
transfusion occur in 1.5∼5.8% of the cases. Most hematomas occur within 12 hours after
sheath removal. Immediate bleeding occurring after sheath insertion is induced by arterial
laceration at the site of the atherosclerotic vessel wall. Otherwise, delayed bleeding after
sheath removal is due to inappropriate compression or coagulation disorders. Clinically,
174 Advances in the Diagnosis of Coronary Atherosclerosis

risk factors are old age, female gender, high blood pressure, use of large sized sheaths, long
procedural times and use of anticoagulants and glycoprotein IIb/IIIa receptor antagonists
before or after procedures. Many arterial closure devices are available as an alternative to
traditional mechanical compression. Although these devices have the potential to reduce the
time to hemostasis, facilitate patient mobilization and decrease hospital length of stay, the
incidence of access-site-related complications is similar or somewhat higher compared with
mechanical compression (Nikolsky et al 2004, Biancari et al 2010). Local bleeding is
transformed to hematoma, which spontaneously resolves in 1∼2 weeks in small hematomas,
or in several weeks or months in large hematomas. Table 4 describes the general
recommendations for postprocedural hemostasis after femoral artery access listed in the
American College of Cardiology/Society for Cardiac Angiography and Interventions
Clinical Expert Consensus Document on Cardiac Catheterization Laboratory Standards
(Bashore et al 2001).

Situation Recommendation
Following hemostatic closure device 1–2 h recumbent in position of comfort,
then ambulate 3 30 min before discharge
Following removal of venous sheath 1 h with leg straight, then ambulate 3 30
min before discharge
Following removal of femoral arterial
sheath (ACT ,175 s)
Manual compression 10–20 min (until hemostasis achieved)
Clamp 15 min to 1 h to achieve hemostasis
Bedrest Leg straight, slight head elevation X 2–6 h
Ambulation 30 min to 1 h before discharge
Table 4. General recommendations regarding postprocedural hemostasis after prior femoral
artery access(Bashore et al. 2001)

3.2 Pseudoaneurysm
After removal of the sheath and compression, the puncture site is surrounded and blocked
by local hematoma. Inappropriate compression favours come-and-go blood flow between
artery lumen and aneurysm surrounded by hematoma. Femoral peudoaneurysm is the
second most common complication occurring in 0.1% to 0.2% of diagnostic angiograms and
3.5% to 5.5% of interventional procedures (Demirbas et al 2005, Kronzon 1997). If hemostasis
is achieved using manual compression, the pseudoaneurysm rate is higher in superficial
femoral artery than common femoral artery access, however rates are similar when using
closure devices (Gutzeit et al 2011). In patients with acute coronary syndrome who are
undergoing emergency angiography and possible intervention, pseudoaneurysm rate
needing closure is lower in radial compared with femoral angiography (HR 0·30, 95% CI
0·13—0·71; p=0·006) (Jolly et al 2011). Small femoral pseudoaneurysm is usually filled with
clots and does not require treatment, however, larger femoral pseudoaneurysms may lead
to secondary complications including rupture, local pain and compression of the adjacent
femoral vein or nerve (Kronzon 1997). Color Doppler ultrasonography is the best diagnostic
tool with a positive diagnostic rate of 95%, however other imaging modalities such as 3-
dimentional CT angiography and angiography are useful. Using color-Doppler
ultrasonography, flow inside the aneurysm sac and the “to-and-fro flow” at the neck of the
lesion is definitely diagnostic. Application of good external compression after sheath
Diagnosis and Management of Complications of Invasive Coronary Angiography 175

removal, selecting a small-bore introducer and sheath removal at a low activated clotting
time (<200 sec) are important factors preventing pseudoaneurysm. Ultrasonography guided
compression of the communicating canal of the pseudoaneurysm stops the blood flow and
leads to clotting and obliteration with a success rate of 84-93% in patients without
anticoagulation (Lange et al 2001, Gabriel et al 2007, Schaub et al 1994). Ultrasound-guided
thrombin injection is another effective means with a success rate of 97-100% (Schneider et al
2009, Weinmann et al 2002). Other endovascular approaches, such as stent graft repair and
coil embolization, are also available. Open surgical repair should be considered after non-
surgical procedure failure.

Fig. 1. Doppler ultrasonography image showing a flow jet from superficial femoral artery to
pseudoaneurysm surrounded by huge hematoma

Fig. 2. Computerized tomographic (CT) findings of pseudoaneurysm in a patient who


underwent coronary angiography. A: Contrast enhanced pseudoaneurysm (arrow) is seen at
anterior aspect of the superficial femoral artery. B: Three-dimensional reconstruction of the
same patient showing saccular pseudoaneurysm (arrow), superficial femoral artery and
anteria profunda femoris.
176 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 3. Angiographic findings of pseudoaneurysm before (A) and after (B) coil embolization.

3.3 Arteriovenous fistula


Although iatrogenic femoral arteriovenous fistula is very rare with an incidence of 0.22%, it
is a significant complication after femoral puncture technique (Sidawy et al 1993). The
fistulas originating at the superficial femoral artery or at the profunda femoris artery drain
to the superficial femoral, profunda femoris vein or its lateral circumflex branch. Below the
femoral artery bifurcation, profunda femoralis vein crosses laterally behind the proximal
superficial femoral artery and then lies in a posterior location to the profunda femoris
artery. Therefore, it is important to avoid a low groin puncture by identifying the level of
the inguinal ligament or localizing the distal half of the femoral head by fluoroscopy
(Sidawy et al. 1993, Lamar R 1990). Early angiography and surgical correction are
recommended for optimal results.

3.4 Retroperitoneal hematoma


Retroperitoneal hematoma occurs in less than 1% of cases of percutaneous coronary
intervention. Clinical signs and symptoms include decreased haematocrit, back or
inguinal pain, abdominal pain, diaphoresis, bradycardia and hypotension. Early
recognition of retroperitoneal hematoma is very important because the diagnosis is often
delayed as symptoms are nonspecific and difficult to recognize because blood silently
tracks in the loose retroperitoneal fatty tissue. Factors predisposing to retroperitoneal
hematoma are female gender, low body surface area, and higher femoral artery puncture
(Farouque et al 2005). It can be easily detected on computerized tomography as an
abnormal, high-density soft-tissue mass originating at the puncture site and contiguous
with the retroperitoneum with resultant compression or distortion of the normal
retroperitoneal structures. There is lack of clinical evidence for the best management of
retroperitoneal haematoma. Inguinal compression is not effective in most cases.
Retroperitoneal hematoma after femoral artery puncture can usually be treated by volume
replacement, correction of coagulopathy and blood transfusion. A small subset of patients
who are unresponsive to volume resuscitation require open repair of bleeding vessel
(Kent et al 1994).
Diagnosis and Management of Complications of Invasive Coronary Angiography 177

Fig. 4. Contrast-enhanced CT scan showing retropelvic hematoma one day after coronary
angiography via right femoral artery

3.5 Spasm
Arterial spasm near the puncture site occurs when arterial size is too small for the sheath.
The transradial approach for coronary angiography or angioplasty is increasingly being
used as an alternative to femoral access due to its low rate of local complications. Arterial
spasm is the most common complication of this technique especially in female patients. Use
of hydrophilic coated sheath, but not long sheath, reduces the incidence of radial artery
spasm during transradial coronary procedures (Rathore et al 2010). Intra-arterial or
subcutaneous infusion of nitroglycerin reduces arterial spasm and facilitates introduction of
catheters (Candemir et al 2009).

4. Conclusion
It is critical to make every attempt at minimizing occurrence of complications during and
after coronary angiography and interventional procedures; beyond operator’s experience
and the patient factors noted, this involves care and attentiveness. The impact of improved
understanding and techniques in intervention should be added to management
considerations. Coronary angiography and intervention is continuously evolving and
knowledge acquired through clinical experience and availability of new devices should
therefore allow safer procedures in the future.

5. References
Ammann P, Brunner-La Rocca HP, Angehrn W, Roelli H, Sagmeister M and Rickli H (2003).
Procedural complications following diagnostic coronary angiography are related to
the operator's experience and the catheter size. Catheter Cardiovasc Interv 59. 13-8.
Barrett BJ and Carlisle EJ (1993). Metaanalysis of the relative nephrotoxicity of high- and
low-osmolality iodinated contrast media. Radiology 188. 171-8.
Bashore TM, Bates ER, Berger PB, Clark DA, Cusma JT, Dehmer GJ, Kern MJ, Laskey WK,
O'Laughlin MP, Oesterle S, Popma JJ, O'Rourke RA, Abrams J, Bates ER, Brodie BR,
Douglas PS, Gregoratos G, Hlatky MA, Hochman JS, Kaul S, Tracy CM, Waters DD
178 Advances in the Diagnosis of Coronary Atherosclerosis

and Winters WL, Jr. (2001). American College of Cardiology/Society for Cardiac
Angiography and Interventions Clinical Expert Consensus Document on cardiac
catheterization laboratory standards. A report of the American College of
Cardiology Task Force on Clinical Expert Consensus Documents. J Am Coll Cardiol
37. 2170-214.
Biancari F, D'Andrea V, Di Marco C, Savino G, Tiozzo V and Catania A (2010). Meta-
analysis of randomized trials on the efficacy of vascular closure devices after
diagnostic angiography and angioplasty. Am Heart J 159. 518-31.
Briguori C, Airoldi F, D'Andrea D, Bonizzoni E, Morici N, Focaccio A, Michev I, Montorfano
M, Carlino M, Cosgrave J, Ricciardelli B and Colombo A (2007). Renal Insufficiency
Following Contrast Media Administration Trial (REMEDIAL): a randomized
comparison of 3 preventive strategies. Circulation 115. 1211-7.
Bruns FJ, Segel DP and Adler S (1978). Control of cholesterol embolization by
discontinuation of anticoagulant therapy. Am J Med Sci 275. 105-8.
Busing KA, Schulte-Sasse C, Fluchter S, Suselbeck T, Haase KK, Neff W, Hirsch JG,
Borggrefe M and Duber C (2005). Cerebral infarction: incidence and risk factors
after diagnostic and interventional cardiac catheterization--prospective evaluation
at diffusion-weighted MR imaging. Radiology 235. 177-83.
Candemir B, Kumbasar D, Turhan S, Kilickap M, Ozdol C, Akyurek O, Atmaca Y and Altin
T (2009). Facilitation of radial artery cannulation by periradial subcutaneous
administration of nitroglycerin. J Vasc Interv Radiol 20. 1151-6.
Delaney A, Carter A and Fisher M (2006). The prevention of anaphylactoid reactions to
iodinated radiological contrast media: a systematic review. BMC Med Imaging 6. 2.
Demirbas O, Batyraliev T, Eksi Z and Pershukov I (2005). Femoral pseudoaneurysm due to
diagnostic or interventional angiographic procedures. Angiology 56. 553-6.
Farouque HM, Tremmel JA, Raissi Shabari F, Aggarwal M, Fearon WF, Ng MK, Rezaee M,
Yeung AC and Lee DP (2005). Risk factors for the development of retroperitoneal
hematoma after percutaneous coronary intervention in the era of glycoprotein
IIb/IIIa inhibitors and vascular closure devices. J Am Coll Cardiol 45. 363-8.
Fine MJ, Kapoor W and Falanga V (1987). Cholesterol crystal embolization: a review of 221
cases in the English literature. Angiology 38. 769-84.
Fukumoto Y, Tsutsui H, Tsuchihashi M, Masumoto A and Takeshita A (2003). The incidence
and risk factors of cholesterol embolization syndrome, a complication of cardiac
catheterization: a prospective study. J Am Coll Cardiol 42. 211-6.
Gabriel M, Pawlaczyk K, Waliszewski K, Krasinski Z and Majewski W (2007). Location of
femoral artery puncture site and the risk of postcatheterization pseudoaneurysm
formation. Int J Cardiol 120. 167-71.
Gruberg L, Mintz GS, Mehran R, Gangas G, Lansky AJ, Kent KM, Pichard AD, Satler LF and
Leon MB (2000). The prognostic implications of further renal function deterioration
within 48 h of interventional coronary procedures in patients with pre-existent
chronic renal insufficiency. J Am Coll Cardiol 36. 1542-8.
Gutzeit A, Graf N, Schoch E, Sautter T, Jenelten R and Binkert CA (2011). Ultrasound-
guided antegrade femoral access: comparison between the common femoral artery
and the superficial femoral artery. Eur Radiol 21(6). 1323-8
Hamon M, Gomes S, Oppenheim C, Morello R, Sabatier R, Lognone T, Grollier G,
Courtheoux P and Hamon M (2006). Cerebral microembolism during cardiac
Diagnosis and Management of Complications of Invasive Coronary Angiography 179

catheterization and risk of acute brain injury: a prospective diffusion-weighted


magnetic resonance imaging study. Stroke 37. 2035-8.
Hong SJ, Wong JT and Bloch KJ (2002). Reactions to radiocontrast media. Allergy Asthma
Proc 23. 347-51.
Jolly SS, Yusuf S, Cairns J, Niemela K, Xavier D, Widimsky P, Budaj A, Niemela M, Valentin
V, Lewis BS, Avezum A, Steg PG, Rao SV, Gao P, Afzal R, Joyner CD, Chrolavicius
S and Mehta SR (2011). Radial versus femoral access for coronary angiography and
intervention in patients with acute coronary syndromes (RIVAL): a randomised,
parallel group, multicentre trial. Lancet 377(9775). 1409-20
Kelly AM, Dwamena B, Cronin P, Bernstein SJ and Carlos RC (2008). Meta-analysis:
effectiveness of drugs for preventing contrast-induced nephropathy. Ann Intern
Med 148. 284-94.
Kent KC, Moscucci M, Mansour KA, DiMattia S, Gallagher S, Kuntz R and Skillman JJ
(1994). Retroperitoneal hematoma after cardiac catheterization: prevalence, risk
factors, and optimal management. J Vasc Surg 20. 905-10; discussion 910-3.
Krause RS (2010). Anaphylaxis: e-Medicine from WebMD
Kronzon I (1997). Diagnosis and treatment of iatrogenic femoral artery pseudoaneurysm: a
review. J Am Soc Echocardiogr 10. 236-45.
Lamar R BR, Rama K. (1990). Femoral arteriovenous fistula as a complication of
percutaneous transluminal coronary angioplasty. A report of five cases. Am Surg
56. 702-6.
Lange P, Houe T and Helgstrand UJ (2001). The efficacy of ultrasound-guided compression
of iatrogenic femoral pseudo-aneurysms. Eur J Vasc Endovasc Surg 21. 248-50.
McCullough PA (2008). Contrat-induced acute kidney injury. J Am Coll Cardiol 51. 1419-28.
Nikolsky E, Mehran R, Halkin A, Aymong ED, Mintz GS, Lasic Z, Negoita M, Fahy M,
Krieger S, Moussa I, Moses JW, Stone GW, Leon MB, Pocock SJ and Dangas G
(2004). Vascular complications associated with arteriotomy closure devices in
patients undergoing percutaneous coronary procedures: a meta-analysis. J Am Coll
Cardiol 44. 1200-9.
Rathore S, Stables RH, Pauriah M, Hakeem A, Mills JD, Palmer ND, Perry RA and Morris JL
(2010). Impact of length and hydrophilic coating of the introducer sheath on radial
artery spasm during transradial coronary intervention: a randomized study. JACC
Cardiovasc Interv 3. 475-83.
Schaub F, Theiss W, Heinz M, Zagel M and Schomig A (1994). New aspects in ultrasound-
guided compression repair of postcatheterization femoral artery injuries.
Circulation 90. 1861-5.
Schneider C, Malisius R, Kuchler R, Lampe F, Krause K, Bahlmann E and Kuck KH (2009). A
prospective study on ultrasound-guided percutaneous thrombin injection for
treatment of iatrogenic post-catheterisation femoral pseudoaneurysms. Int J Cardiol
131. 356-61.
Scolari F, Bracchi M, Valzorio B, Movilli E, Costantino E, Savoldi S, Zorat S, Bonardelli S,
Tardanico R and Maiorca R (1996). Cholesterol atheromatous embolism: an
increasingly recognized cause of acute renal failure. Nephrol Dial Transplant 11.
1607-12.
Sidawy AN, Neville RF, Adib H and Curry KM (1993). Femoral arteriovenous fistula
following cardiac catheterization: an anatomic explanation. Cardiovasc Surg 1. 134-7.
180 Advances in the Diagnosis of Coronary Atherosclerosis

Solomon R and Dauerman HL (2010). Contrast-induced acute kidney injury. Circulation 122.
2451-5.
Sutton AG FP, Grech ED, Hall JA, Stewart MJ, Davies A, de Belder MA (2001). Early and late
reactions after the use of iopamidol 340, ioxaglate 320, and iodixanol 320 in cardiac
catheterization. Am Heart J 141. 677-83.
Weinmann EE, Chayen D, Kobzantzev ZV, Zaretsky M and Bass A (2002). Treatment of
postcatheterisation false aneurysms: ultrasound-guided compression vs
ultrasound-guided thrombin injection. Eur J Vasc Endovasc Surg 23. 68-72.
Wilson DM, Salazer TL and Farkouh ME (1991). Eosinophiluria in atheroembolic renal
disease. Am J Med 91. 186-9.
10

Coronary Angiography and


Contrast-Induced Nephropathy
Omer Toprak
Balikesir State Hospital, Department of Nephrology, Balikesir
Turkey

1. Introduction
Radiologic procedures such as coronary angiography utilizing intravascular contrast media
(CM) injections are being widely applied for diagnostic and therapeutic purposes. These led
to a parallel increase in the incidence of contrast-induced nephropathy (CIN). Renal failure
requiring dialysis after contrast media administration is associated with a 40% in-hospital
mortality rate and has a 2-year mortality rate of 80%. Approximately $180 million are spent
annually to manage CIN in the US. The large number of patients who are severely affected
by CIN underscores the importance of addressing known risk factors and preventions for
CIN. Patients at risk for CIN can often be identified with a routine medical history, physical
examination and laboratory analysis before the procedure. Two of the major risk factors of
CIN are pre-existing renal failure and diabetes mellitus. After the high-risk patient
population has been identified and risk factors addressed, the next step in preventing CIN is
the use of different prophylactic therapies. The following chapter provides an overview of
current risk factors and preventive methods of CIN.

2. Definition, differential diagnosis and incidence of CIN


The most commonly used definition for CIN in clinical trial is the elevation of serum
creatinine by ≥0.5mg/dl or ≥25% occurring within 48 hours after administration of CM, and
the absence of an alternative etiology. Serum creatinine is the standard marker to detect
CIN. However, serum creatinine demonstrates major limitations. Using the Cockcroft-Gault
and the Modification of Diet in Renal Disease equations are useful in estimation of the GFR.
Serum cystatin C has been proposed as an alternative endogenous marker of GFR showing
higher correlation to standard clearance methods such as inulin or iohexol clearance. Serum
cystatin C may detect CIN one to two days earlier than creatinine. Also, recent studies
documented that serum and urine neutrophil gelatinase-associated lipocalin is an early
predictive biomarker of CIN in patients underwent coronary angiography (Shaker et al.,
2010). Cholesterol atheroemboli, volume depletion, and interstitial nephritis should consider
in differential diagnosis of CIN. Before coronary angiography, the volume status of high-
risk patients can be assessed through the inferior vena cava index or bioimpedance
spectroscopy. The incidence of CIN is reported to be 0.6-2.3% in general population who do
not have any risk factor for CIN, but the incidence can be increased to 90% in patients at
high risk for CIN (Toprak, 2007).
182 Advances in the Diagnosis of Coronary Atherosclerosis

2.1 Pathophysiology of CIN


The pathogenesis of CIN is unclear. The proposed mechanisms are medullary hypoxia due
to decreased renal blood flow secondary to renal artery vasoconstriction, tubular obstruction
and direct tubular toxicity of the CM due to apoptosis and oxidative damage. Endothelial
dysfunction and renal microcirculatory alterations also may play a role. The development of
CIN is affected by changes in renal hemodynamics, such as increased activity of renal
vasoconstrictors and decreased activity of renal vasodilators. Other factors that may
decrease renal blood flow include increased viscosity of CM and increased erythrocyte
aggregation induced by CM, which results in diminished oxygen delivery. Contrast agents
have been found to reduce antioxidant enzyme activity, and direct cytotoxic effects
mediated by oxygen free radicals. Apoptosis is also involved as a result of cellular injury.
Hyperosmolar contrast agents induce renal hemodynamic changes caused by osmolar-
driven solute diuresis with activation of tubuloglomerular feedback or an increase in tubular
hydrostatic pressures, which may cause compression of the intrarenal microcirculation and a
decreased GFR. Also hyperosmolar contrast agents have direct toxic effects on renal
epithelial cells. In addition, DNA fragmentation was increased in cells exposed to
hyperosmolar CM.

2.2 Clinical course and outcomes


CIN may range in severity from asymptomatic, nonoliguric transient renal dysfunction to
oliguric severe renal failure that necessitates permanent dialysis. CIN is reported to be the
third leading cause of in-hospital acute renal failure after hypotension and surgery.
Approximately $180 million is spent annually to manage CIN in the US. Dangas et al.
showed that in-hospital outcomes such as death (6.3% vs 0.8%), cardiac death (4.0% vs
0.5%), coronary artery bypass grafting (5.8% vs 0.5%), major adverse cardiac event (9.3% vs
1.1%), packed red cell transfusion (28% vs 6%), vascular surgery of access site (5.6% vs
2.6%), post-procedure length of stay (6.8±7.1 vs 2.3±2.5) were significantly higher in CIN
developed patients compare with control (p<0.0001). In cumulative one-year outcome death,
out-of-hospital death and major adverse cardiac events were significantly higher in CIN
developed patients (p<0.0001) (Dangas et al., 2005). In a study of acute myocardial infarction
patients undergoing primary angioplasty, it was found that CIN developed patients have
significantly higher incidence of high-rate atrial fibrillation (p=0.01), high-degree conduction
disturbances requiring permanent pacemaker (p=0.04), acute pulmonary edema (p=0.008),
respiratory failure requiring mechanical ventilation (p<0.0001), cardiogenic shock requiring
intra-aortic balloon (p<0.0001), and acute renal failure requiring renal replacement therapy
(p<0.0001) ( Marenzi et al., 2004).

2.3 Risk factors for CIN


Specific factors that increase the risks for development of CIN are related to the patient, the
contrast media, and the procedure (Table 1).

2.3.1 Patient-related risk factors


Older age
The possible reasons of the high incidence of CIN in elderly were age-related changes in
renal function, more difficult vascular access following tortuosity, calcification of the vessels
Coronary Angiography and Contrast-Induced Nephropathy 183

Risk Factors Odds Ratio (95%CI) p Value


Kidney Related Risk Factors
Pre-existing renal failure
Preprocedural creatinine 2.0-2.9 mg/dl 7.37 (4.78-11.39) <0.0001
Preprocedural creatinine ≥ 3mg/d 12.82 (8.01-20.54) <0.0001
Diabetes mellitus-Diabetic nephropathy
Preprocedural creatinine≤ 1.1mg/dl 1.86 (1.20-2.89) 0.005
Preprocedural creatinine 1.2-1.9 mg/dl 2.42 (1.54-3.79) <0.001
Use of nephrotoxic drugs
Low effective circulatory volume 1.19 (0.72-1.95) 0.05
Cardiovascular System Related Risk Factors
Class III-IV congestive heart failure 2.20 (1.60-2.90) <0.0001
Left ventricle ejection fraction<40% 1.57 (1.14-2.16) 0.005
Acute myocardial infarction ≤ 24 h 1.85 (1.31-2.63) 0.0006
Hypertension 2.00 (1.40-2.80) 0.0001
Periprocedural hypotension 2.50 (1.70-3.69) <0.00001
Multi-vessel coronary involvement 3.24 (1.07-9.82) 0.038
Peripheral vascular disease 1.90 (1.40-2.70) <0.0001
Preprocedure shock 1.19 (0.72-1.96) 0.05
Using intra-aortic balloon pump 15.51 (4.65-51.64) <0.0001
Bypass graft intervention 4.94 (1.16-20.9) 0.03
Time-to-reperfusion ≥6 h 2.51 (1.01-6.16) 0.04
Pulmonary edema 2.56 (1.42-4.52) 0.001
Demographic Risk Factors
Age >75 years 5.28 (1.98-14.05) 0.0009
Female gender 1.4 (1.25-1.60) 0.0001
Contrast Media Related Risk Factors
High total dose of contrast agent (>300 ml) 2.8 (1.17-6.68) 0.02
Osmolality (Low- vs. high-osmolality) 0.50 (0.36-0.68)
Short duration of two contrast administration 4.4 (2.9-6.5) <0.0001
Other Possible Risk Factors
Procedural success 0.27 (0.19-0.38) <0.0001
Baseline hematocrit 0.95 (0.92-0.97) <0.00001
Hyperuricemia 4.71 (1.29-17.21) 0.019
ACE inhibitors 3.37 (1.14-9.94) 0.028
Angiotensin Receptor Blockers 2.70 (1.25-5.81) 0.011
Metabolic Syndrome 426 (1.19-15.25) 0.026
Hypoalbuminemia 5.79 (1.71-19.64) 0.005
Hypercholesterolemia
Renal transplant
Multiple myeloma
Diuretics
Intra-arterial contrast administration
Sepsis, cirrhosis
Table 1. Risk factors for the development of contrast-induced nephropathy
184 Advances in the Diagnosis of Coronary Atherosclerosis

requiring greater amount of CM, defective prostaglandin synthesis, and the presence of
renovascular disease. Furthermore, hypovolemia is very common in elderly patients. In a
prospective study in which elderly patients (≥70 years) were subjected to cardiac
catheterization, 11% developed CIN (Rich & Crecelius, 1990). In another study, CIN
incidence was 17% in elderly patients (>60 years) as compared with 4% in younger patients
(Kohli et al., 2000). In 208 patients with acute myocardial infarction who underwent
coronary intervention, it was found that an age of ≥75 years was an independent risk for
CIN (OR=5.28, p=0.0009) (Marenzi et al., 2004).
Gender
In a retrospective study of 8628 patients who underwent PCI, female sex was an
independent predictor of CIN (OR=1.4, p<0.0001). One-year outcome analyses by gender
showed a higher mortality among females than among males in a cohort of CIN patients
(14% vs 10%, p=0.05) (Iakovou et al., 2003). The findings of this study contradict those of a
previous randomized controlled trial of ionic vs nonionic CM, in which a multivariate
analysis identified male gender as an independent risk factor for CIN (Rudnick et al., 1995).
Whether female gender is truly an independent predictor of CIN will require further
confirmatory studies.
Pre-existing renal disease
The major risk factor for CIN is a GFR<60 ml/min/1.73 m2. Chronic kidney disease is
associated with decreased vasodilatory response, which is important in developing CIN,
and in patients with renal insufficiency, the clearance of CM is slower than in normal
subjects. In a study of 7586 patients who underwent coronary intervention, CIN developed
in 22.4% of the patients who had serum creatinine levels of 2.0 to 2.9 mg/dl and in 30.6% of
those with serum creatinine levels of 3.0 mg/dl or higher, compared with 2.4% of patients
with serum creatinine levels <1.1 mg/dl ( Rihal et al., 2002). Two other studies (Moore et al.,
1992; Barrett et al., 1992) reported that the incidence of CIN increased from 4% to 20% as the
baseline serum creatinine increased from 1.2 to 2.9 mg/dl. In another study, the incidence of
CIN increased from 8% to 92% as the serum creatinine increased from 1.5 to 6.8 mg/dl.
Furthermore, the probability of CIN requiring dialysis increases from 0.04% to 48% as the
baseline GFR decreases from 50 to 10 ml/min (McCullough et al., 1997).
Diabetes mellitus
Patients with diabetes constitute an important group at high risk of developing CIN.
Patients with diabetic nephropathy and a mean serum creatinine of 6.8 mg/dl had a 92%
incidence of CIN after coronary angiography (Weinrauch et al., 1977). Patients with diabetes
who have advanced chronic renal failure because of causes other than diabetic nephropathy
are at significantly higher risk of developing CIN like diabetic nephropathy. On the other
hand, studies have shown that when pre-existing renal disease is present, patients with and
without diabetes are similarly at risk of CIN, which correlates with the degree of renal
disease. Some authors have suggested that DM in the absence of nephropathy, particularly
in insulin-dependent patients with diabetes, is associated with an increased risk of CIN
(McCullough et al., 1997; Toprak 2007). In a study, it was found that the incidence of CIN
was rather low (2%) in patients with neither diabetes nor azotemia, significantly higher
(16%) in individuals with diabetes but preserved renal function, and much higher (38%) in
patients who had both diabetes and azotemia (Lautin et al., 1991). In another study, the
incidence of CIN was found to be 2% in patients without diabetes and 3.7% in patients with
Coronary Angiography and Contrast-Induced Nephropathy 185

diabetes with a baseline creatinine of 1.1 mg/dl or less (OR=1.86, p=0.005). When renal
function is mildly impaired (serum creatinine level 1.2 to 1.9 mg/dl), the risk of CIN in
patients with diabetes mellitus increases to 4.5% (OR=2.42, p<0.001) ( Rihal et al., 2002).
Other studies have failed to corroborate this connection (Parfrey et al., 1989). However,
given that, those with diabetes alone were found to be at slightly higher risk of CIN than the
general population.
Pre-diabetes
In a study of 421 patients who underwent coronary angiography with renal insufficiency,
we presented that pre-DM increase the incidence of CIN 2.1-fold in comparison to patients
with normal fasting glucose (NFG) but pre-DM is not as strong as DM as a risk of
developing CIN. CIN occurred in 20% of the DM (RR=3.6, p=0.001), 11.4% of the pre-DM
(RR 2.1, p=0.314) and 5.5% of the NFG group. The decrease of GFR was higher in DM and
pre-DM (p=0.001 and p=0.002, respectively). Length of hospital stay was 2.45 ± 1.45 day in
DM, 2.27 ± 0.68 day in pre-DM, and 1.97 ± 0.45 day in NFG (p<0.001, DM vs. NFG and
p=0.032, pre-DM vs. NFG). The rate of major adverse cardiac events was 8.7% in DM, 5% in
pre-DM, and 2.1% in NFG (P=0.042, DM vs. NFG). Hemodialysis was required in 3.6% of
DM, and 0.7% in pre-DM (P=0.036, DM vs. NFG), and the total number of hemodialysis
sessions during 3 months was higher in DM and pre-DM (P<0.001). Serum glucose ≥124
mg/dl was the best cut-off point for prediction of CIN (Toprak et al., 2007).
Congestive heart failure and reduced left ventricular ejection fraction
Studies have shown that reduced left ventricular ejection fraction (LVEF) (≤49%) and
advanced congestive heart failure are independent risk factors for CIN. In a study, Dangas
et al. showed that LVEF below 40% is an independent predictor of CIN (Dangas et al., 2005).
We have previously reported that if the LVEF is greater than 30%, this condition does not
show any significant effect on the development of CIN (Toprak et al., 2003). In a study it was
shown that congestive heart failure was an independent risk for CIN (OR=1.53, p=0.007)
(Rihal et al., 2002). In a cohort study it was found that congestive heart failure is a risk for
CIN in patients who underwent PCI (OR=2.2, p<0.0001) (Bartholomew et al., 2004).
Hypertension
Hypertension has been categorized as a risk factor for CIN in some research. In a study of
8628 patients who underwent percutaneous interventions, hypertension was found to be an
independent predictor of CIN (OR=1.2, p=0.0035) (Iakovou et al., 2003). In another cohort
study, hypertension was a risk for CIN in patients who underwent PCI (OR=2.0, p=0.0001)
(Bartholomew et al., 2004).
ACE Inhibitors and angiotensin receptor blockers
ACE inhibitors have been identified as a risk factor for CIN because of their potential to
reduce renal function. On the other hand, some small studies have shown that the
nephrotoxicity of CM may be reduced because of decreased renal vasoconstriction by
inhibition of angiotensin II. In a randomized controlled study with 71 patients with diabetes
who underwent coronary angiography randomized to captopril or control, 25-mg captopril
was given three times daily. There was a significant decrease in CIN in the patients who
received captopril compared with the control group (6% vs 29%, respectively, p<0.02)
(Gupta et al., 1999). We have performed a randomized controlled study in 80 patients with
serum creatinine below 2 mg/dl who underwent coronary angiography. Captopril was
186 Advances in the Diagnosis of Coronary Atherosclerosis

administered in 48 patients before coronary angiography. Five patients (10.4%) in the


captopril group developed CIN, compared with only one patient (3.1%) in the control group
(p=0.02) (Toprak et al., 2003). In a study of 230 patients with renal insufficiency and age ≥65
years we found that chronic ACE inhibitor administration was a risk for developing CIN.
CIN occurred in 17 patients (15.6%) in the ACE inhibitor group and 7 patients (5.8%) in the
control group (p=0.015). Chronic ACE inhibitor administration was a risk indicator of CIN
(OR=3.37, p=0.028) (Cirit et al., 2006). In another study, 421 patients with renal insufficiency
who underwent coronary angiography, use of ACE inhibitors or ARB was a risk for CIN in
multivariate analysis (OR=2.7, p=0.011) (Toprak et al., 2007). In a recent study, the impact of
renin-angiotensin and aldosterone system blockade on the frequency of CIN was assessed
retrospectively. Patients treated with ACE inhibitors or ARB (n=269) and were not treated
with them (n=143) underwent coronary angiography included to the study. CIN developed
11.9% in ACE-inhibitor using group and 4.2% in control group (p=0.006). Use of ARB or
ACE inhibitors was found as a risk for CIN (OR=3.08, p=0.016) (Kiski et al., 2010). Checking
the use of ACE inhibitors or ARB before coronary angiography seems to be a useful guide in
tracking risk assessment for CIN.
Nephrotoxic drugs
Sulfonamides, aminoglycosides, and their combination with furosemide are particularly
potent. Cyclosporin A may intensify medullary hypoxia, and cisplatin can attach to
sulfhydryl groups. Mannitol can increase the metabolic workload in the kidney, and
amphotericin B can cause the effect of a combination of mannitol and cyclosporine A.
However, the individual roles of these medications as independent risk factors of CIN have
not been determined in large trials. Nonetheless, it seems quite likely that nonselective
NSAIDs and selective COX-2 inhibitors decrease the vasodilatory prostaglandins in the
kidney and potentiate the vasoconstrictive effect of CM.
Multiple myeloma
The pathomechanism of this process has been explained by the precipitation of CM molecules
together with Tamm–Horsfall proteins and other abnormal proteins, tubular epithelial cells
damaged and desquamated as a result of ischemia, direct contrast toxicity, or disturbed
function of integrins. In studies conducted on animals, intratubular light chains, particularly in
the setting of intravascular volume depletion, have been found to augment the nephrotoxic
potential of CM (Holland et al., 1985). Studies showed an incidence of CIN of only 0.6–1.25%
in patients with myeloma if dehydration is avoided (McCarthy & Becker, 1992).
Metformin
The oral antidiabetic agent metformin is not itself nephrotoxic, but it is known that patients
who are receiving metformin may develop lactic acidosis as a result of CIN. A decline in
renal function after contrast exposure could adversely affect the clearance of metformin. The
complication was almost always observed in diabetic patients with decreased renal function
before injection of CM. It seems safer to instruct patients especially at high risk for CIN not
to take this drug for 48 h or so after CM administration and resume taking the drug only if
there are no signs of nephrotoxicity.
Hypercholesterolemia
In the literature, there are limited studies on the relationship between hypercholesterolemia
and CIN. According to these studies, hypercholesterolemia aggravates CIN through the
Coronary Angiography and Contrast-Induced Nephropathy 187

reduced production of nitric oxide (Yang et al., 2004). Altered nitric oxide-dependent renal
vasodilatation, which is important in the pathogenesis of CIN, is prevalent in
hypercholesterolemia.
Hyperuricemia
It has been suggested that tubular obstruction by uric acid plays a role in the pathogenesis
of CIN. In a prospective cohort study we evaluated 266 patients who undergoing elective
coronary angiography and we found that patients with hyperuricemia are at risk of
developing CIN (OR=4.71, p=0.019). CIN occurred in 15.1% of the hyperuricemic group and
2.9% of the normouricemic group (p<0.001). Length of hospital stay (p<0.001) and CIN
requiring renal replacement therapy (p=0.017) were significantly higher in hyperuricemic
group. Serum uric acid 7 mg/dl in males and 5.9 mg/dl in females were found the best
cut-off value for prediction of CIN (Toprak et al., 2006).
Multivessel coronary involvement, peripheral vascular disease, and renal artery stenosis
Factors related to accelerated or diffuse atherosclerosis are linked to the development of
CIN. The treatment of multivessel disease, challenging chronic total occlusions and
extensively diseased coronary segments, may require high doses of CM for providing an
optimal image quality, thus enhancing the potential toxic effects on the renal function. If a
patient has multivessel coronary involvement, the other vessels in the body, such as the
renal artery, can be involved. In a study of 177 patients who underwent cardiac
catheterization, subjects were also evaluated for renal artery stenosis. Coronary artery
disease was detected in 110 patients (62%), and significant renal artery stenosis was detected
in 19 patients (11%). Using multivariate analysis, it was found that the extent of coronary
artery disease was an independent predictor of renal artery stenosis (Weber-Mzell et al.,
2002). In a study a total of 5571 patients who underwent PCI were evaluated for CIN risk
factors, and it was found that multivessel coronary involvement was only a univariate
predictor of CIN (p=0.003) (Mehran et al., 2004). In two other cohort studies it was found
that peripheral vascular disease is a risk for CIN in patients who underwent PCI (OR=1.9,
p<0.0001 and OR=1.71, p=0.001, respectively) (Bartholomew et al., 2004; Rihal et al., 2002).
In a study a total of 219 non-diabetic patients who underwent coronary angiography we
have found that multivessel coronary involvement is a risk for CIN (OR=3.24, p=0.038)
(Toprak et al., 2006).
Hypovolemia
Decreased effective circulating volume and reduced renal perfusion potentiate renal
vasoconstriction after administration of intravascular CM. The toxic effects of CM on the
renal tubular lumen may be exacerbated in hypovolemia. At present the most convincing
preventive procedure of CIN is adequate hydration with isotonic saline or sodium
bicarbonate, and intravenous hydration seems to have better results than does oral
hydration. Before angiography, the volume status of patients can be assessed through the
inferior vena cava index, mean atrial pressure, noninvasive pulmonary-capillary wedge
pressure or bioimpedance spectroscopy (Toprak & Cirit, 2005).
Renal transplantation
Patients with renal transplantation may be at a higher risk of CIN due to concomitant use of
cyclosporine and higher prevalence of diabetes and renal insufficiency. In a study, 33
patients with a functioning renal allograft who underwent different contrast studies, the
incidence of CIN was 21.2% (Ahuja et al., 2000).
188 Advances in the Diagnosis of Coronary Atherosclerosis

Acute myocardial infarction


A study by Rihal et al. showed that acute myocardial infarction within 24 h before
administration of the CM is a risk factor for CIN (OR=1.85, p=0.0006). This study
demonstrates that CIN is a frequent complication in acute myocardial infarction, even in
patients with a normal baseline renal function. (Rihal et al., 2002). In a study of 208 acute
myocardial infarction patients who underwent primary PCI, anterior acute myocardial
infarction was significantly higher in patients who developed CIN (p=0.0015). However, in
multivariate analysis, anterior acute myocardial infarction (OR=2.17, p=0.09) was not a risk
for CIN (Marenzi et al., 2004). In 2082 percutaneous interventions for acute myocardial
infarction, it was reported a more than seven-fold (3.2% vs 23.3%) increase in 1-year
mortality in patients who developed CIN (Sadeghi et al., 2003).
Low hematocrit level
A baseline hematocrit value of less than 39% for men and less than 36% for women is a risk
for CIN. The relationship between low hematocrit levels and CIN has been investigated in a
prospective study of 6773 patients who underwent PCI (Nikolsky et al., 2005). A lower
baseline hematocrit was an independent predictor of CIN; and each 3% decrease in baseline
hematocrit resulted in a significant increase in the odds of CIN in patients with and without
chronic kidney disease (11% and 23%, respectively). Dangas et al. showed that the baseline
hematocrit level is an independent predictor of CIN in patients with chronic kidney disease
(OR=0.95, p<0.00001) (Dangas et al., 2005).
Low serum albumin
Hypoalbuminemia impairs endothelial function, enhances renal vasoconstriction, impairs
the synthesis and release of nitric oxide, and decreases antioxidant enzyme activity. In a
study, low serum albumin (<3.5 g/dl) was identified as a risk factor for CIN in patients 70
years of age or older who underwent cardiac catheterization (Rich, et al., 1990). Also we
have found that in 230 patients who underwent coronary angiography with renal
insufficiency, serum albumin level ≤3.5 g/dl was a risk factor for CIN (OR=5.79, p=0.005)
(Cirit et al., 2006).
Hypotension, sepsis, cirrhosis, and pulmonary edema
A systolic blood pressure of less than 80 mm Hg for at least 1 h that requires inotropic
support with medications is a risk factor for CIN. A study by Dangas et al showed that
periprocedural hypotension and pulmonary edema are independent predictors of CIN in
patients with chronic kidney disease (OR=2.50, p<0.00001 and OR=2.56, p=0.001,
respectively) (Dangas et al., 2005) Sepsis, through direct damage by bacterial toxins to renal
tubules and impairment of circulation, has also been reported as a risk factor. Reduction of
effective intravascular volume caused by liver cirrhosis has been reported as contributing to
pre-renal reduction in renal perfusion, thus enhancing the ischemic insult of CM (Toprak,
2007).
Metabolic syndrome, impaired fasting glucose and hypertriglyceridemia
In a prospective cohort study of 219 non-diabetic elderly patients with reduced kidney
function who underwent elective coronary angiography, we reported that metabolic
syndrome was a risk indicator of CIN (OR=4.26, p=0.026). CIN occurred in 14% of the
metabolic syndrome group and 3.6% of the non-metabolic syndrome group (relative risk
3.93, p=0.007). Impaired fasting glucose (OR=4.72, p=0.007), high triglyceride (OR=4.06,
Coronary Angiography and Contrast-Induced Nephropathy 189

p=0.022); and multi-vessel involvement (OR=3.14, p=0.038) in the metabolic syndrome


group were predictors of CIN (Toprak et al., 2006).

2.3.2 Procedure-related risk factors


Short duration of the two contrast administration and urgent/emergency procedure
In those who have no risk factors for CIN, angiography should be delayed more than 48
hours after a previous exposure to intravascular contrast media. In patients with diabetes or
preexisting renal disease, this time interval should be increased to more than 72 hours. In a
cohort study, urgent/emergency procedure was found as a predictor of CIN (OR=4,
p<0.0001) (Bartholomew et al., 2004). The higher risk of developing CIN in patients with
urgent status was irrespective of baseline renal function.
Use of intra-aortic balloon pump
In 208 consecutive acute myocardial infarction patients undergoing percutaneous coronary
intervention, use of intra-aortic balloon pump was a risk predictor of CIN (OR=15.51,
p<0.0001) (Marenzi et al., 2004). In a study, it has demonstrated that, intra-aortic balloon
pump use is an independent predictor of CIN in patients with chronic kidney disease
(OR=2.27, p=0.004) (Dangas et al., 2005). In another study, it was found that the use of intra-
aortic balloon pump was a risk factor for CIN requiring dialysis after PCI (OR=1.94)
(Gruberg et al., 2001). In another derivation and validation cohort study, intra-aortic balloon
pump use was a risk for CIN in patients undergoing coronary intervention (OR=5.1,
p<0.0001) (Bartholomew et al., 2004).
Bypass graft intervention and delayed reperfusion
Procedures with bypass angiography and intervention may be associated with higher
complexity, longer duration, and limited success, thus indicating an unstable post-
procedural period with impaired cardiac output. Gruberg et al. showed that the risk of CIN
requiring dialysis after PCI was increased with bypass graft intervention (OR=4.94)
(Gruberg et al., 2001). In a study of 208 acute myocardial infarction patients undergoing
primary PCI, the risk of CIN was increased if the time-to-reperfusion is ≥6 h (OR=2.51,
p=0.04) (Marenzi et al., 2004)

2.3.3 Contrast medium-related risk factors


Increased dose of contrast medium
According to different sources, the relatively safe cutoff point of contrast amount varies
from 70 ml up to 220ml. However, doses as low as 20 to 30 ml are capable of inducing CIN.
In a study that patients undergoing coronary angiography, each 100 ml of contrast medium
administered was associated with a significant increase of 12% in the risk of CIN (OR=1.12,
p=0.02) (Rihal et al., 2002). Marenzi et al. showed that contrast volume >300 ml is an
independent risk for CIN (OR=2.80, p=0.02) (Marenzi et al., 2004). In another study patients
with preexisting renal failure revealed a 10-fold risk of CIN when more than 125 ml of
contrast media was administered (Taliercio et al., 1986).
High-osmolar and ionic CM
In a large study which comparing the non-ionic low-osmolality agent iohexol to the ionic
high-osmolality agent meglumine/sodium diatrizoate in patients with pre-existing renal
190 Advances in the Diagnosis of Coronary Atherosclerosis

dysfunction undergoing angiography, patients with renal insufficiency receiving diatrizoate


were 3.3 times as likely to develop CIN compared to those receiving iohexol (Rudnick et al.,
1995). NEPHRIC trial is a randomized, prospective study comparing the nonionic iso-
osmolar CM iodixanol with the nonionic low-osmolar CM iohexol in 129 renal impairment
patients with diabetes undergoing coronary or aorto-femoral angiography. The incidence of
CIN was 3% in the iodixanol group and 26% in the iohexol group (p=0.002) (Aspelin et al.,
2003). In another randomized study, the renal tolerance of iodixanol and iohexol was
compared in 124 patients with creatinine >1.7 mg/dl. The incidence of CIN was 3.7% in
iodixanol group and 10% in iohexol group (p>0.05) (Chalmers et al., 1999). The available
data do not provide clear evidence that the whole iso-osmolar CM class offers an
improvement over the low-osmolar CM class. Other studies with iodixanol in renal failure
patients have shown a higher incidence of CIN than that observed in the NEPHRIC study
(21% in the RAPPID trial, 30% in the CONTRAST trial) (Baker et al., 2003: Stone et al., 2003).
In addition to their osmolarity, contrast medias are characterized as ionic versus non-ionic.
Small clinical trials of low-risk patients undergoing coronary angiography have shown little
difference in the risk of CIN between the 2 types of CM. However, a randomized trial of
1196 patients undergoing coronary angiography showed that non-ionic CM reduced the
incidence of CIN in patients with preexisting renal disease with or without diabetes
(Rudnick et al., 1995). In addition, symptomatic or hemodynamic adverse drug events have
been shown to occur less often with non-ionic, low-osmolality CM compared with ionic,
high-osmolality CM. In high-risk patients, it is reasonable to don’t use the high-osmolar and
ionic CM to minimize the risk of CIN.
Intra-arterial administration of the contrast media
Intra-arterial contrast administration is a risk for CIN. This effect is thought to be due to the
fact that the acute intra renal concentration of CM is much higher after intra arterial rather
than intravenous injection.

2.3.4 Scoring method to predict high risk patients for CIN


Mehran et al. developed a simple scoring method that integrates eight baseline clinical
variables to assess the risk of CIN after percutaneous coronary intervention (PCI). These are
hypotension (score 5), use of intra-aortic balloon pump (score 5), congestive heart failure
(score 5), serum creatinine>1.5 mg/dl (score 4), age>75 years (score 4), anemia (score 3),
diabetes mellitus (score 3), and volume of CM (score 1 per 100 ml). If the total score is 5 or
less, the risk category is low; if the total score is 16 or higher, the risk category is very high
(Mehran et al., 2004)

2.4 Prevention strategies for CIN


Extracellular volume expansion with intravenous saline or sodium bicarbonate, minimizing
the dose of CM, using low-osmolar non-ionic CM instead of high osmolar ionic CM,
stopping the intake of nephrotoxic drugs and avoiding short intervals between procedures
requiring CM have all been shown to be effective in reducing CIN. Alternatives to ordinary
CM, such as carbon dioxide or gadolinium chelates, can be used in patients at high risk of
CIN (Table 2).
Volume expansion
Volume expansion is the single most important measure that has been documented to be
beneficial in preventing CIN. A standardized saline hydration protocol has been proven
Coronary Angiography and Contrast-Induced Nephropathy 191

Clinical evidence advocating With conflicting or


Don’t use
their use limited evidence
Nonsteroidal anti-
Extracellular volume inflammatory drugs, COX-2
Acetylcystein
expansion inhibitors, aminoglycoside,
cisplatin
Saline or sodium bicarbonate Loop diuretics Theophylline
Low or iso-osmolar contrast Mannitol Calcium channel blockers
Minimizing the dose of Multiple use of contrast
Fenoldopam
contrast within 72 h
Alternative imaging
Large doses of contrast Captopril
techniques
Monitoring serum creatinine High-osmolar contrast Ascorbic acid
Delaying contrast procedures
Metformin usage especially in
until hemodynamic status is Atrial natriuretic peptide
patients with renal failure
corrected
≥48 h between contrast
Endothelin antagonist
procedures
PGE1
Hemofiltration
Nebivolol
Statins
B-type natriuretic peptide
Pentoxifylline
Table 2. Prevention strategies for contrast-induced nephropathy in high-risk patients
effective in reducing the risk of CIN and should be used routinely. The most widely
accepted protocol is administering isotonic saline at 1 to 1.5 ml/kg/h beginning 6 to 12
hours prior to the procedure and continuing for up to 12 hours following
contrastadministration. In a randomized trial, two different hydration regimens were
compared in 1620 patients undergoing coronary interventions. They showed that the
incidence of CIN was significantly lower among patients given an isotonic saline solution
than among those given a hypotonic saline solution (0.7% vs. 2.0% respectively, p=0.04)
(Mueller et al., 2002). In another trial, a total of 119 patients with serum creatinine exceeding
1.1 mg/dl were randomized to receive isotonic solution of sodium bicarbonate (n=59) or
isotonic saline (n=60) at a rate of 3 ml/kg/h for 1 hour before and 1 ml/kg/h for 6 hours
after contrast administration. CIN developed in only 1 patient (1.7%) compared with 8
patients (13.6%) in the saline group (p=0.02) (Merten et al., 2004). The authors postulated
that a reduction in oxidative injury may have conferred protection against CIN. However,
further studies are required to clarify the role of hydration with sodium bicarbonate in
preventing CIN. In a prospective study, the effect of combination intravenous and oral
volume supplementation on the development of CIN was studied in 425 patients
undergoing percutaneous coronary intervention. Patients were randomly assigned to
receive hydration with either isotonic or half-isotonic. In addition patients were encouraged
to drink plenty of fluids (at least 1500 ml). They found that applying the combination of
intravenous and oral volume supplementation results in a very low incidence of CIN (1.4%)
192 Advances in the Diagnosis of Coronary Atherosclerosis

(Mueller et al., 2005). Most studies have found that hydration alone is better than hydration
combined with a diuretic. In a study, 78 patients with serum creatinine >1.6 mg/dl were
randomized to three groups: hydration alone, hydration with mannitol and hydration with
furosemide. Half-isotonic saline was used for hydration. CIN occurred in 11%, 28% and 40%
of patients in the three groups, respectively (p=0.02), thus showing that forced diuresis is of
no benefit in preventing CIN. In a meta-analysis it was found that the administration of
sodium bicarbonate is superior to the administration of saline alone in the prevention of
CIN (Solomon et al., 1994). However, further controlled clinical trials are needed due to
significant study heterogeneity and publication bias. The effectiveness of sodium
bicarbonate treatment to prevent CIN in high-risk patients remains uncertain.
N-acetylcysteine
It had been postulated that antioxidant N-acetylcysteine (NAC) might scavenge oxygen free
radicals, thus attenuate the cytotoxic effects of CM. NAC may also have direct vasodilating
effects on the kidneys through an increase in the biologic effects of nitric oxide. Tepel et al.
were evaluated the effects of NAC (600 mg orally twice daily), at first time, in 83 patients
undergoing computed tomography. Two percent of the patients in the NAC group had CIN
versus 21% in the placebo group (p=0.01) (Tepel et al., 2000). Since then, a number of trials
have been published. Results from these trials have been inconsistent. In a randomized,
placebo-controlled study it was found that NAC is protective against CIN Fifty-four patients
were randomized to receive either 600 mg of NAC twice daily for 4 doses or placebo. The
incidence of CIN was 8% in the NAC group versus 45% in the placebo group (p=0.005)
(Diaz-Sandoval et al., 2002). In addition to oral administration, intravenous administration
of NAC to protect against CIN has also been evaluated. In a study, Baker et al. randomly
assigned 80 patients to receive either NAC infusion (n=41) versus saline infusion (n=39).
CIN developed in only 2 (5%) of patients in the NAC group compared with 8 (21%) in the
saline group (p=0.04) (Baker et al., 2003). The authors concluded that NAC infusion protects
against CIN. In a meta-analysis, evaluating more than 800 patients at high risk of
developing CIN also documented a positive impact of NAC prophylaxis on CIN (Birck et
al., 2003). In another meta-analysis, nine randomized controlled trials were included and the
difference in mean change in creatinine between the NAC treated group and controls was -
0.27 mg/dl. The relative risk of developing CIN was 0.43 in subjects randomized to NAC.
They suggest that NAC helps prevent declining renal function and CIN (Liu et al., 2005). In
contrast to these reports, some studies failed to find a significant effect of NAC on the
occurrence of CINA total of 183 patients with preexisting renal insufficiency undergoing
contrast study were randomly assigned to receive NAC at a dose of 600 mg twice daily on
the day before and the day of the contrast study plus saline infusion or saline alone. The
incidence of CIN was 6.5% in the NAC group versus 11% in the control group (p=0.22)
(Briguori et al., 2002). In a multi centric double blind clinical trial 156 patients undergoing
coronary angiography or percutaneous coronary intervention with creatinine clearance <50
ml/min were randomly assigned to receive N-acetylcysteine 600 mg orally twice daily for
two days or placebo. Sixteen patients developed CIN. Eight of 77 patients (10.4%) in the
NAC group and eight of 79 patients (10.1%) in the placebo group (p=1.00). No difference
was observed in the change in endogenous creatinine clearance, p=0.28). They concluded
that oral NAC did not prevent CIN in patients at low to moderate risk undergoing cardiac
catheterisation with ionic low osmolality CM (Gomes et al., 2005). In another study, 50
patients undergoing elective diagnostic coronary angiography with serum creatinine values
Coronary Angiography and Contrast-Induced Nephropathy 193

above 1.3 mg/dl were included and CIN was detected in 3 of 25 patients (12%) in the NAC
group and 2 of 25 patients (8%) in the control group (p>0.05). It was detected that in patients
planned to undergo elective diagnostic coronary angiography with renal dysfunction, oral
NAC and hydration before the procedure was not more effective than hydration alone in the
prevention of CIN (Gulel et al., 2005). A direct renoprotective effect of NAC remains
questionable. To date, only a few trials described the effects of NAC not only on serum
creatinine but also on clinical end points. The serum creatinine can be decrease in
administration of NAC without renoprotective effect. In a prospective study, NAC was
given at a dose of 600 mg every 12 h for a total of four doses to the volunteers with a normal
renal function who did not receive contrast agent. There was a significant decrease of the
mean serum creatinine (p<0.05) and a significant increase of the GFR (p<0.02), whereas the
cystatin C concentration did not change significantly (Hoffmann et al., 2004). In patients
undergoing emergency diagnostic procedures, in whom a full hydration protocol is not
possible, an abbreviated hydration regimen plus oral or intravenous administration of NAC
can be recommended. NAC may be of benefit mostly in high-risk patients. If NAC is to be
used as a preventative measure, it should be given at a dose of 600 mg orally twice daily on
the day before and day of the procedure.
Ascorbic acid
Prophylactic oral administration of ascorbic acid may protect against CIN. In a randomized,
placebo-controlled trial in 231 patients with serum creatinine concentration ≥1.2 mg/dl who
undergoing coronary angiography showed that the use of ascorbic acid was associated with
a significant reduction in the rate of CIN. CIN occurred in 11 of the 118 patients (9%) in the
ascorbic acid group and in 23 of the 113 patients (20%) in the placebo group (OR=0.38;
p=0.02) (Spargias et al., 2004). Further prospective studies are needed to validate these
preliminary results.
Fenoldopam
Fenoldopam mesylate is a selective dopamine-1 receptor agonist that produces systemic,
peripheral and renal arterial vasodilatation. Several investigators have reported a positive
impact of fenoldopam against CIN in small studies. In a placebo-controlled, double-blind,
multicenter trial, 315 patients with creatinine clearance of less than 60 ml/min were
randomized to receive fenoldopam infusion [0.05 µg/kg/min titrated to 0.1 µg/kg/min
(n=157)] or matching placebo (n=158). CIN occurred in 33.6% of patients in the
fenoldopam group compared with 30.1% of patients in the placebo group (p=0.61) (Stone
et al., 2003). The authors concluded that fenoldopam did not protect against CIN. In 2
other large studies comparing fenoldopam with NAC treatment with fenoldopam either
had a similar, non significant effect as that of NAC or was inferior to it (Allaqaband et al.,
2002; Briguori et al., 2004). Therefore, the routine use of fenoldopam cannot be
recommended at the present time.
Adenosine antagonists
CM stimulate the intrarenal secretion of adenosine, which binds to the renal adenosine
receptor and acts as a potent vasoconstrictor, reducing renal blood flow and increasing the
generation of oxygen free radicals as it is metabolized to xanthine and hypoxanthine.
Theophylline and aminophylline, adenosine antagonists, have also been studied in the
prevention of CIN in a number of trials. Studies with these agents have used varying doses
and dosage forms and yielded conflicting results (Erley et al., 1999; Kapoor et al., 2001).
194 Advances in the Diagnosis of Coronary Atherosclerosis

Based on the conflicting information found in clinical studies, adenosine antagonists should
not be routinely used in patients as a preventative measure at this time.
Calcium channel blockers
The calcium channel antagonists verapamil and diltiazem have been found to attenuate the
renal vasoconstrictor response after exposure to CM. However, when the efficacy of the
felodipine, nitrendipine and nifedipine was evaluated, results were inconsistent. Two small
studies performed the use of sublingual nifedipine given prior to contrast administration.
Patients (n=20) who received sublingual nifedipine did not have a significant increase in
serum creatinine, while those in the placebo group did (Rodicio et al., 1990). In another
study, patients (n=30) who received nifedipine had an increase in renal plasma flow
following administration of contrast, while patients in the placebo group had a decrease in
renal flow (Russo et al., 1990). One other study showed that nitrendipine use cause a
significant reduction in the GFR in the placebo group compared to little or no change in GFR
in the nitrendipine group (Neumayer et al., 1989). In another study, 27 patients with normal
to moderately reduced renal function underwent femoral angiography randomized to
receive either oral felodipine or placebo. Patients in the felodipine group had a significant
increase in serum creatinine from baseline, while patients in the placebo group did not
demonstrate a similar increase (Spangberg-Viklund et al., 1996). More large-scale trials are
needed before calcium channel blockers can be routinely recommended in patients prior to
contrast administration.
Prostaglandin E1
PGE1 has vasodilatory effects that may be beneficial in preventing CIN. In one study, 130
patients were randomly assigned to receive either placebo or one of three doses of PGE1. The
increase in serum creatinine level was smaller in all of the three PGE1 groups than in the
placebo group, but the difference was significant only in the medium-dose (20 ng/kg/min)
of PGE1 group (Koch et al., 2000). More studies need to be done to better understand the role
of prostaglandin E1, but results from this pilot study appear promising.
Atrial natriuretic peptide (ANP)
ANP may prevent CIN by increasing renal blood flow. In a study, ANP was included in one
of the four arms. In which dopamine, mannitol, and ANP caused an increase in CIN in
diabetic patients as compared to saline alone (Weisberg et al., 1994). In another trial patients
were randomized to one of four treatment arms: fluid alone or one of three doses of ANP.
Results showed no statistically significant differences in the incidence of CIN between any of
the four treatment arms (Kurnik et al., 1998) Based on these results and the limited clinical
data, ANP cannot be advocated in the prevention of CIN.
Endothelin antagonists
Endothelin-1 is a potent endogenous vasoconstrictor, is thought to play a role in the
development of CIN. Endothelin-1 has two primary receptors. In animal studies, endothelin-
A antagonists were shown to reduce the incidence of CIN (Liss et al., 2003). However, in a
randomized study of 158 patients, the use of a mixed endothelin-A and B antagonist was
associated with a significantly higher incidence of CIN than was placebo (56% vs. 29%,
p=0.002) (Wang et al., 2000). Endothelin antagonists currently have no role in prevention of
CIN.
Coronary Angiography and Contrast-Induced Nephropathy 195

Low-dose of dopamine
At low doses (1-3 mcg/kg/min), dopamine activates two types of dopamine (DA) receptors,
DA-1 and DA-2. Activation of the DA-1 receptor results in an increase in natriuresis and
renal blood flow. Since dopamine, at low doses, is believed to be more selective for the DA-1
receptors, it has been investigated in the prevention of CIN. Kapoor et al. randomized 40
patients with diabetes scheduled to undergo a coronary angiography to either dopamine or
placebo control. None of the patients in the dopamine group developed CIN compared to
50% of patients receiving placebo (Kapoor et al., 2002). In another prospective, randomized
trial, Hans et al. evaluated 55 patients (40% had diabetes) with chronic renal insufficiency.
Patients were randomized to receive dopamine or an equal volume of saline. The group
receiving dopamine had a significantly lower incidence of CIN as compared to the control
group (Hans et al., 1998). In contrast to the trials showing a potential benefit of dopamine,
other studies have failed to demonstrate this benefit. Abizaid et al. performed a randomized,
prospective study involving patients with renal insufficiency who underwent coronary
angioplasty. Patients were randomized to continue with the saline, receive aminophylline in
addition to the saline, or receive dopamine plus saline. In the dopamine plus saline group,
50% of patients developed CIN, while only 30% of the patients in the saline-alone group
developed CIN. This difference did not reach statistical significance, but it appeared that use
of dopamine might worsen outcomes (Abizaid et al., 1999). Low-dose dopamine use cannot
be supported at this time.
Statins
Whether additional benefits can be achieved with the use of statin in decreasing the risk of
CIN remains undetermined. In a recent meta analysis of randomised controlled trials
comparing statin pretreatment with non-statin pretreatment for the prevention of CIN, it
was found that, the incidence of CIN was not significantly lower in statin pretreatment
group as compared with control group (RR=0.76, p=0.30) (Zhang et al., 2011). The current
cumulative evidence suggests that statin pretreatment may neither prevent CIN nor reduce
the need for renal replacement therapy.
Hemofiltration and hemodialysis
Currently available data do not support use of prophylactic hemodialysis for prevention of
CIN. In a trial of 113 patients, reported that CIN occurred in 24% of the hemodialysis group
as compared with 16% of non-hemodialysis group (Vogt et al., 2001). Clinically relevant
events also were not different in two groups. Only continuous venovenous hemofiltration
has been shown to protect against CIN. In a study, 114 patients with chronic renal failure
undergoing percutaneous coronary intervention were divided in two groups: 56 patients
received normal saline and 58 patients underwent hemofiltration at a rate of 1000 ml/h
(Marenzi et al., 2003). Hemofiltration seems to have a protective effect, including significant
reduction in in-hospital and 1-year mortality compared with routine hydration. The
mechanisms of this benefit are not clear. Further studies are needed to confirm the results of
this trial.
New types of contrast medias
Gadolinium-enhanced magnetic resonance coronary angiography is a non-invasive method
for evaluation of coronary arteries. It has been suggested that gadolinum-based CM could
be used in stead of iodinated CM for radiological examinations in patients with significant
196 Advances in the Diagnosis of Coronary Atherosclerosis

renal impairment. However, its use has been questioned on the basis of reports of
nephrotoxicity and its association with nephrogenic systemic fibrosis, a rare and serious
syndrome that involves fibrosis of skin, joints, eyes, and internal organs. In a study by
Hoffmann et al. the effect of gadopentetate dimeglumine (iodine-based CM) was studied in
181 patients with normal renal function and the effect of gadolinium was studied in 198
patients with pre-excisting renal failure. There was no statistically significant change in
serum creatinine concentration after gadopentetate dimeglumine. In contrary, serum
creatinine levels decreased significantly after the administration of gadolinium (p<0.01)
(Hoffmann et al., 2005). In a retrospective study, the safety of gadolinium was evaluated in
91 patients with stage 3 and 4 renal failure who underwent angiographic MRI procedures.
Eleven of 91 patients developed CIN (12.1%) (Ergun et al., 2006). In another randomized
study gadobutrol, a gadolinium-based CM, was compared with standard iohexol, an
iodinated CM, in 21 patients with renal dysfunction. The incidence of CIN was 50% in
gadobutrol group and 45% in iohexol group (p=0.70). In this study, gadolinium showed no
benefit over iohexol in patients with severely impaired renal function (Erley et al., 2004).
More studies need to be done to better understand the role of gadolinum on CIN.
Ultrasound contrast agents are micro-bubbles which produce acoustic enhancement. They
are pharmacologically almost inert and safe.

3. Conclusion
The development of CIN is associated with adverse outcomes including prolonged
hospitalization, the potential need for renal replacement therapy, and most important,
increased mortality. The treatment of established CIN is limited to supportive measures and
dialysis. For this reason, screening for high-risk patients before CM including cardiac
procedures and taking the appropriate prophylactic regimens is important in reducing CIN.
Pre-existing renal dysfunction, especially when secondary to diabetic nephropathy, is the
most important risk factor. Extra cellular volume expansion and use of low osmolar CM are
the two most effective measures to prevent CIN. Acetylcysteine may use in high-risk
patients, but this finding has not been uniform or always demonstrated by currently
available trials.

4. References
Abizaid, AS., Clark, CE., Mintz, GS., Dosa, S., Popma, JJ., Pichard, AD., Satler, LF., Harvey,
M., Kent, KM., & Leon, MB. (1999). Effects of Dopamine and Aminophylline on
Contrast-Induced Acute Renal Failure after Coronary Angioplasty in Patients with
Preexisting Renal Insufficiency. The American Journal of Cardiology, Vol.83, No.2,
(January 1999), pp.260-263, ISSN 0002-9149
Ahuja, TS., Niaz, N., & Agraharkar, M. (2000). Contrast-Induced Nephrotoxicity in Renal
Allograft Recipients. Clinical Nephrology, Vol.54, No.1, (July 2000), pp.11-14, ISSN
0301-0430
Allaqaband, S., Tumuluri, R., Malik, AM., Gupta, A., Volkert, P., Shalev, Y., & Bajwa, TK. (2002).
Prospective Randomized Study of N-Acetylcysteine, Fenoldopam, and Saline for
Prevention of Radiocontrast-Induced Nephropathy. Catheterization and Cardiovascular
Interventions, Vol.57, No.3, (November 2002), pp.279-283, ISSN 1522-1946
Coronary Angiography and Contrast-Induced Nephropathy 197

Aspelin, P., Aubry, P., Fransson, SG., Strasser, R., Willenbrock, R., & Berg, KJ. (2003). Nephrotoxic
Effects in High-Risk Patients Undergoing Angiography. The New England Journal of
Medicine, Vol.348, No.6, (February 2003), pp.491-499, ISSN 0028-4793
Baker, CS., Wragg, A., Kumar, S., De Palma, R., Baker, LR., & Knight, CJ. (2003). A Rapid
Protocol for the Prevention of Contrast-Inducted Renal Dysfunction: The RAPPID
Study. Journal of the American College of the Cardiology, Vol. 41, No. 12, (June 2003),
pp.2114-2118, ISSN 0735-1097
Bartholomew, BA., Harjai, KJ., Dukkipati, S., Boura, JA., Yerkey, MW., Glazier, S., Grines,
CL., & O'Neill, WW. (2004). Impact of Nephropathy after Percutaneous Coronary
Intervention and a Method for Risk Stratification, The American Journal of Cardiology,
Vol.93, No.12, (June 2004), pp.1515-1519, ISSN 0002-9149
Barrett, BJ., Parfrey, PS., Vavasour HM., McDonald, J., Kent, G., Hefferton, D., O'Dea, F.,
Stone, E., Reddy, R., & McManamon, PJ. (1992). Contrast Nephropathy in Patients
with Impaired Renal Function: High Versus Low Osmolar Media. Kidney
International, Vol.41, No.5, (May 1992), pp. 1274–1279, ISSN 0085-2538
Birck, R., Krzossok, S., Markowetz, F., Schnulle, P., van der Woude, FJ., & Braun, C. (2003).
Acetylcysteine for Prevention of Contrast Nephropathy: Meta-Analysis. Lancet,
Vol.362, No.9384, (August 2003), pp.598-603, ISSN 0140-6736
Briguori, C., Manganelli, F., Scarpato, P., Elia, PP., Golia, B., Riviezzo, G., Lepore, S., Librera,
M., Villari, B., Colombo, A., & Ricciardelli, B. (2002). Acetylcysteine and Contrast
Agent-Associated Nephrotoxicity. Journal of the American College of the Cardiology,
Vol.40, No.2, (July 2002), pp. 298-303, ISSN 0735-1097
Briguori, C., Colombo, A., Airoldi, F., Violante, A., Castelli, A., Balestrieri, P., Paolo Elia, P.,
Golia, B., Lepore, S., Riviezzo, G., Scarpato, P., Librera, M., Focaccio, A., &
Ricciardelli, B. (2004). N-Acetylcysteine Versus Fenoldopam Mesylate to Prevent
Contrast Agent-Associated Nephrotoxicity. Journal of the American College of the
Cardiology, Vol.44, No.4, (August 2004), pp.762-765, ISSN 0735-1097
Chalmers, N., & Jackson, RW. (1999). Comparison of Iodixanol and Iohexol in Renal
Impairment. The British Journal of Radiology, Vol.72, No.859, (July 1999), pp.701-703,
ISSN 0007-1285
Cirit, M., Toprak, O., Yesil, M., Bayata, S., Postaci, N., Pupim, L., & Esi, E. (2006).
Angiotensin-Converting Enzyme Inhibitors as a Risk Factor for Contrast-Induced
Nephropathy. Nephron Clinical Practice, Vol.104, No.1, (August 2006), pp. c20-c27,
ISSN 1660-2110
Dangas, G., Iakovou, I., Nikolsky, E., Aymong, ED., Mintz, GS., Kipshidze, NN., Lansky,
AJ., Moussa, I., Stone, GW., Moses, JW., Leon, MB., & Mehran, R. (2005). Contrast-
Induced Nephropathy After Percutaneous Coronary Interventions in Relation to
Chronic Kidney Disease and Hemodynamic Variables. The American Journal of
Cardiology, Vol.95, No.1, (January 2005), pp. 13-19, ISSN 0002-9149
Diaz-Sandoval, LJ., Kosowsky, BD., & Losordo, DW. (2002). Acetylcysteine to Prevent
Angiography-Related Renal Tissue Injury (The APART Trial). American Journal of
Cardiology, Vol.89, No.3, (February 2002), pp.356-358, ISSN 0002-9149
Ergun, I., Keven, K., Uruc, I., Ekmekci, Y., Canbakan, B., Erden, I., & Karatan, O. (2006). The
Safety of Gadolinium in Patients with Stage 3 and 4 Renal Failure. Nephrology
Dialysis Transplantation, Vol.21, No.3, (March 2006), pp. 697-700, ISSN 0931-0509
Erley, CM., Bader, BD., Berger, ED., Tuncel, N., Winkler, S., Tepe, G., Risler, T., & Duda, S.
Gadolinium-Based Contrast Media Compared with Iodinated Media for Digital
Subtraction Angiography in Azotaemic Patients. Nephrology Dialysis
Transplantation, Vol.19, No.10, (October 2004), pp.2526-2531, ISSN 0931-0509
198 Advances in the Diagnosis of Coronary Atherosclerosis

Erley, CM., Duda, SH., Rehfuss, D., Scholtes, B., Bock, J., Muller, C., Osswald, H., & Risler, T.
(1999). Prevention of Radiocontrast Media-Induced Nephropathy in Patients with
Pre-Existing Renal Insufficiency by Hydration in Combination with the Adenosine
Antagonist Theophylline. Nephrology Dialysis Transplantation, Vol.14, No.5, (May
1999), pp.1146–1149, ISSN 0931-0509
Gomes, VO., Poli de Figueredo, CE., Caramori, P., Lasevitch, R., Bodanese, LC., Araujo, A.,
Roedel, AP., Caramori, AP., Brito, FS Jr., Bezerra, HG., Nery, P., & Brizolara, A. (2005).
N-Acetylcysteine Does Not Prevent Contrast Induced Nephropathy after Cardiac
Catheterisation with an Ionic Low Osmolality Contrast Medium: A Multicentre Clinical
Trial. Heart, Vol.91, No.6, (June 2005), pp.774-778, ISSN 1355-6037
Gruberg, L., Mehran, R., Dangas, G., Mintz, GS., Waksman, R., Kent, KM., Pichard, AD.,
Satler, LF., Wu, H., & Leon, MB. (2001). Acute Renal Failure Requiring Dialysis
after Percutaneous Coronary Interventions. Catheterization and Cardiovascular
Interventions, Vol.52, No.4, (April 2001), pp.409-416, ISSN 1522-1946
Gulel, O., Keles, T., Eraslan, H., Aydogdu, S., Diker, E., & Ulusoy, V. (2005). Prophylactic
Acetylcysteine Usage for Prevention of Contrast Nephropathy after Coronary
Angiography. Journal of Cardiovascular Pharmacology, Vol.46, No.4, (October 2005),
pp.464-467, ISSN 0160-2446
Gupta, RK., Kapoor, A., Tewari, S., Sinha, N., & Sharma, RK. (1999). Captopril for Preventing of
Contrast-Induced Nephropathy in Diabetic Patients: A Randomized Study. Indian Heart
Jouurnal, Vol.51, No.5, (September 1999), pp.521–526, ISSN 0019-4832
Hans, SS., Hans, BA., Dhillon, R., Dmuchowski, C., & Glover, J. (1998). Effect of Dopamine
on Renal Function after Arteriography in Patients With Preexisting Renal
Insufficiency. The American Surgeon, Vol.34, No.5, (May 1998), pp.1682-1688, ISSN
0003-1348
Hoffmann, U., Fischereder, M., Reil, A., Fischer, M., Link, J., & Kramer, BK. (2005). Renal
Effects of Gadopentetate Dimeglumine in Patients with Normal and Impaired
Renal Function. European Journal of Medical Research, Vol. 10, No.4, (April 2005),
pp.149-154, ISSN 0949-2321
Hoffmann, U., Fischereder, M., Kruger, B., Drobnik, W.; & Kramer, BK. (2004). The Value of
N-Acetylcysteine in the Prevention of Radiocontrast Agent-Induced Nephropathy
Seems Questionable. Journal of the American Society of Nephrology, Vol.15, No.2,
(February 2004), pp.407–410, ISSN 1046-6673
Holland, MD., Galla, JH., Sanders, PW., & Luke, RG. (1985). Effect of Urinary pH and
Diatrizoate on Bence Jones Protein Nephrotoxicity in the Rat. Kidney International,
Vol.27, No.1, (January 1985), pp. 46-50, ISSN 0085-2538
Iakovou, I., Dangas, G., Mehran, R., Lansky, AJ., Ashby, DT., Fahy, M., Mintz, GS., Kent,
KM., Pichard, AD., Satler, LF., Stone, GW., & Leon, MB. (2003). Impact of Gender
on the Incidence and Outcome of Contrast-Induced Nephropathy after
Percutaneous Coronary Intervention. The Journal of Invasive Cardiology, Vol.15, No.1,
(January 2003), pp. 18–22, ISSN 1042-3931
Kapoor, A., Kumar, S., Gulati, S., Gambhir, S., Sethi, RS., & Sinha, N. The Role of Theophylline in
Contrast-Induced Nephropathy: A Case-Control Study. Nephrology Dialysis
Transplantation, Vol. 17, No.11, (November 2002), pp.1936–1941, ISSN 0931-0509
Kapoor, A., Sinha, N., Sharma, RK., Shrivastava, S., Radhakrishnan, S., Goel, PK., & Bajaj R.
(1996). Use of Dopamine in Prevention of Contrast Induced Acute Renal Failure: A
Randomized Study. International Journal of Cardiology,Vol.53, No.3, (March 1996),
pp.233-236, ISSN 0167-5273
Coronary Angiography and Contrast-Induced Nephropathy 199

Kiski, D., Stepper, W., Brand, E., Breithardt, G., & Reinecke, H. (2010). Impact of Renin-
Angiotensin-Aldosterone Blockade by Angiotensin-Converting Enzyme Inhibitors
or AT-1 Blockers on Frequency of Contrast Medium-Induced Nephropathy: A Post-
Hoc Analysis from the Dialysis-Versus-Diuresis (DVD) Trial. . Nephrology Dialysis
Transplantation, Vol.25, No.3, (May 2010), pp.759-64, ISSN 0931-0509
Koch, JA., Plum, J., Grabensee, B., & Modder, U. (2000). Prostaglandin E1: A New Agent for
the Prevention of Renal Dysfunction in High Risk Patients Caused by
Radiocontrast Media? Nephrology Dialysis Transplantation, Vol. 15, No.1, (January
2000), pp.43-49, ISSN 0931-0509
Kohli, HS., Bhaskaran, MC., Muthukumar, T., Thennarasu, K., Sud, K., Jha, V., Gupta, KL.,
& Sakhuja, V. (2000). Treatment-Related Acute Renal Failure in the Elderly: A
Hospital-Based Prospective Study. . Nephrology Dialysis Transplantation, Vol.15,
No.2, (February 2000), pp. 212-217, ISSN 0931-0509
Kurnik, BR., Allgren, RL., Genter, FC., Solomon, RJ., Bates, ER., & Weisberg, LS. (1998).
Prospective Study of Atrial Natriuretic Peptide for the Prevention of Radiocontrast-
Induced Nephropathy. American Journal of the Kidney Diseases, Vol.31, No.4, (April
1998), pp.674-680, ISSN 0272-6386
Lautin, EM., Freeman, NJ., Schoenfeld, AH., Bakal, CW., Haramati, N., Friedman, AC.,
Lautin, JL., Braha, S., Kadish, EG., & Sprayregen, S. (1991). Radiocontrast-
Associated Renal Dysfunction: Incidence and Risk Factors. AJR American Journal of
Roentgenology, Vol.157, No.1, (July 1991), pp. 49–58, ISSN 0033-8419
Liss, P., Carlsson, PO., Nygren, A., Palm, F., & Hansell, P. (2003). ET-A Receptor Antagonist
BQ123 Prevents Radiocontrast Media-Induced Renal Medullary Hypoxia. Acta
Radiologica, Vol.44, No.1, (January 2003), pp.111-117, ISSN 0284-1851
Liu, R., Nair, D., Ix, J., Moore, DH., & Bent, S. (2005). N-Acetylcysteine for the Prevention of
Contrast-Induced Nephropathy. A Systematic Review and Meta-Analysis. Journal of
General Internal Medicine, Vol.20, No.2, (Febrary 2005), pp.193-200, ISSN 0884-8734
McCarthy, CS., & Becker, JA. (1992). Multiple Myeloma and Contrast Media. Radiology,
Vol.183, No.2, (May 1992), pp.519–521, ISSN 0033-8419
McCullough, PA., Wolyn, R., Rocher, LL., Levin, RN., & O'Neill, WW. (1997). Acute Renal
Failure after Coronary Intervention: Incidence, Risk Factors, and Relationships to
Mortality. The American Journal of Medicine, Vol.103, No.5, (November 1997), pp.
368–375, ISSN 0002-9243
Marenzi, G., Lauri, G., Assanelli, E., Campodonico, J., De Metrio, M., Marana, I., Grazi, M.,
Veglia, F., & Bartorelli, AL. (2004). Contrast-Induced Nephropathy in Patients
Undergoing Primary Angioplasty for Acute Myocardial Infarction. Journal of the
American College of the Cardiology, Vol.44, No.9, (November 2004), pp. 1780-1785,
ISSN 0735-1097
Marenzi, G., Marana, I., Lauri, G., Assanelli, E., Grazi, M., Campodonico, J., Trabattoni, D.,
Fabbiocchi, F., Montorsi, P., & Bartorelli, AL. (2003). The Prevention of
Radiocontrast-Agent-Induced Nephropathy by Hemofiltration. The New England
Journal of Medicine, Vol.349, No.14, (October 2003), pp.1333-1340, ISSN 0028-4793
Mehran, R., Aymong, ED., Nikolsky, E., Lasic, Z., Iakovou, I., Fahy, M., Mintz, GS., Lansky,
AJ., Moses, JW., Stone, GW., Leon, MB., & Dangas, G. (2004). A Simple Risk Score
for Prediction of Contrast-Induced Nephropathy after Percutaneous Coronary
Intervention: Development and Initial Validation. Journal of the American College of
Cardiology. Vol.44, No.7, (October 2004), pp.1393-1399, ISSN 0735-1097
Merten, GJ., Burgess, WP., Gray, LV., Holleman, JH., Roush, TS., Kowalchuk, GJ., Bersin,
RM., Van Moore, A., Simonton, CA 3rd., Rittase, RA., Norton, HJ., & Kennedy, TP.
200 Advances in the Diagnosis of Coronary Atherosclerosis

(2004). Prevention of contrast-induced nephropathy with sodium bicarbonate: a


randomized controlled trial. Journal of the American Medical Association, Vol.291,
No.19, (May 2004), pp.2328-2334, ISSN 0098-7484
Moore, RD., Steinberg, EP., Powe, NR., Brinker, JA., Fishman, EK., Graziano, S., & Gopalan,
R. (1992). Nephrotoxicity of High-Osmolarity vs Low-Osmolarity Contrast Media:
Randomized Clinical Trial. Radiology, Vol.182, No.3, (March 1992), pp. 649–655,
ISSN 0033-8419
Mueller, C., Seidensticker, P., Buettner, HJ., Perruchoud, AP., Staub, D., Christ, A., &
Buerkle, G. (2005). Incidence of Contrast Nephropathy in Patients Receiving
Comprehensive Intravenous and Oral Hydration. Swiss Medical Weekly, Vol.135,
No.19, (May 2005), pp.286-290, ISSN 1424-7860
Mueller, C., Buerkle, G., Buettner, HJ., Petersen, J., Perruchoud, AP., Eriksson, U., Marsch,
S., & Roskamm, H. (2002). Prevention of Contrast Media-Associated Nephropathy:
Randomized Comparison of 2 Hydration Regimens In 1620 Patients Undergoing
Coronary Angioplasty. Archives of Internal Medicine, Vol.162, No.3, (February 2002),
pp.329-336, ISSN 0003-9926
Neumayer, HH., Junge, W., Kufner, A., & Wening, A. (1989). Prevention of Radiocontrast-
Media-Induced Nephrotoxicity by the Calcium Channel Blocker Nitrendipine: A
Prospective Randomized Clinical Trial. Nephrology Dialysis Transplantation, Vol.4,
No.12, (April 1989), pp.1030-1036, ISSN 0931-0509
Nikolsky, E., Mehran, R., Lasic, Z., Mintz, GS., Lansky, AJ., Na, Y., Pocock, S., Negoita, M.,
Moussa, I., Stone, GW., Moses, JW., Leon, MB., & Dangas, G. Low Hematocrit
Predicts Contrast-Induced Nephropathy After Percutaneous Coronary
Interventions. Kidney International, Vol.67, No.2, (February 2005), pp.706-713, ISSN
0085-2538
Parfrey, PS., Griffiths, SM., Barrett, BJ., Paul, MD., Genge, M., Withers, J. Farid, N., &
McManamon, PJ. (1989). Contrast Material-Induced Renal Failure in Patients with
Diabetes Mellitus, Renal Insufficiency, or Both. A Prospective Controlled Study. The
New England Journal of Medicine, Vol.320, No.3, (January 1989), pp.143-149, ISSN
0028-4793
Rich, MW., & Crecelius, CA. (1990). Incidence, Risk Factors, and Clinical Course of Acute
Renal Insufficiency after Cardiac Catheterization in Patients 70 Years of Age or
Older. A Prospective Study. Archives of Internal Medicine, Vol.150, No.6, (June1990),
pp. 1237-1242), ISSN 0003-9926
Rihal, CS., Textor, SC., Grill, DE., Berger, PB., Ting, HH., Best, PJ., Singh, M., Bell, MR., Barsness,
GW., Mathew, V., Garratt, KN., & Holmes, DR Jr. (2002). Incidence and Prognostic
Importance of Acute Renal Failure after Percutaneous Coronary
Intervention. Circulation, Vol.105, No.105, (May 2002), pp. 2259-2264, ISSN 0009-7322
Rodicio, JL., Morales, JM., & Ruilope, LM. (1990). Calcium Antagonists and the Kidney.
Nephrology Dialysis Transplantation, Vol.5, No.2, (1990), pp.81-86, ISSN 0931-0509
Rudnick, MR., Goldfarb, S., Wexler, L., Ludbrook, PA., Murphy, MJ., Halpern, EF., Hill, JA.,
Winniford, M., Cohen, MB., & Van Fossen, DB. (1995). Nephrotoxicity of Ionic and
Nonionic Contrast Media in 1196 Patients: A Randomized Trial. The Iohexol
Cooperative Study. Kidney International, Vol.47, No.1, (January 1995), pp. 254-261,
ISSN 0085-2538
Russo, D., Testa, A., Della Volpe, L., & Sansone, G. (1990). Randomised Prospective Study
on Renal Effects of Two Different Contrast Media in Humans: Protective Role of a
Calcium Channel Blocker. Nephron, Vo.55, No.3, (1990), pp.254-257, ISSN 0028-2766
Coronary Angiography and Contrast-Induced Nephropathy 201

Sadeghi, HM., Stone, GW., Grines, CL., Mehran, R., Dixon, SR., Lansky, AJ., Fahy, M., Cox,
DA., Garcia, E., Tcheng, JE., Griffin, JJ., Stuckey, TD., Turco, M., & Carroll, JD.
(2003). Impact of Renal Insufficiency in Patients Undergoing Primary Angioplasty
for Acute Myocardial Infarction. Circulation, Vol.108, No.22, (December 2003),
pp.2769-2775, ISSN 0009-7322
Shaker, OG., El-Shehaby, A., & El-Khatib, M. (2010). Early Diagnostic Markers for Contrast
Nephropathy in Patients Undergoing Coronary Angiography. Angiology, Vol.61,
No.8, (November 2010), pp. 731-736, ISSN 0003-3197
Solomon, R., Werner, C., Mann, D., D’Elia, J., & Silva, P. (1994). Effects of Saline, Mannitol
and Furosemide to Prevent Acute Decreases in Renal Function Induced by
Radiocontrast Agents. The New England Journal of Medicine, Vol. 331, No.21,
(November 1994), pp.1416–1420, ISSN 0028-4793
Spangberg-Viklund, B., Berglund, J., Nikonoff, T., Nyberg, P., Skau, T., & Larsson, R. (1996).
Does Prophylactic Treatment with Felodopine, a Calcium Antagonist, Prevent Low-
Osmolar Contrast Induced Renal Dysfunction in Hydrated Diabetic and
Nondiabetic Patients with Normal or Moderately Reduced Renal Function?
Scandinavian Journal of Urology and Nephrology, Vol.30, No.1, (February 1996), pp.63-
68, ISSN 0036-5599
Spargias, K., Alexopoulos, E., Kyrzopoulos, S., Iokovis, P., Greenwood, DC., Manginas, A.,
Voudris, V., Pavlides, G., Buller, CE., Kremastinos, D., & Cokkinos, DV. (2004).
Ascorbic Acid Prevents Contrast-Mediated Nephropathy in Patients with Renal
Dysfunction Undergoing Coronary Angiography or Intervention. Circulation,
Vol.110, No.18, (November 2004), pp.2837-2842, ISSN 0009-7322
Stone, GW., McCullough, PA., Tumlin, JA., Lepor, NE., Madyoon, H., Murray, P., Wang, A.,
Chu, AA., Schaer, GL., Stevens, M., Wilensky, RL., & O'Neill, WW., CONTRAST
Investigators. (2003). Fenoldopam Mesylate for the Prevention of Contrast-Induced
Nephropathy: A Randomized Controlled Trial. The Journal of the American Medical
Association, Vol.290, No.17, (November 2003), pp.2284-2291, ISSN 0098-7484
Taliercio, CP., Vlietstra, RE., Fisher, LD., & Burnett, JC. (1986). Risks for Renal Dysfunction
with Cardiac Angiography. Annals of Internal Medicine, Vol.104, No.4, (April 1986),
pp.501-504, ISSN 0003-4819
Tepel, M., van Der Giet, M., Schwarzfeld, C., Laufer, U., Liermann, D., & Zidek, W. (2000).
Prevention of Radiographic-Contrast-Agent-Induced Reductions in Renal Function
by Acetylcysteine. The New England Journal of Medicine, Vol.343, No.3, (July2000),
pp.,180-184, ISSN 0028-4793
Toprak, O., Cirit, M., Tanrisev, M., Yazici, C., Canoz, O., Sipahioglu, M., Uzum, A., Ersoy,
R., & Sozmen, EY. (2008). Preventive Effect of Nebivolol on Contrast-Induced
Nephropathy in Rats. Nephrology Dialysis Transplantation, Vol.23, No.3, (March
2008), pp. 853-859, ISSN 0931-0509
Toprak, O. (2007). Conflicting and New Risk Factors for Contrast-Induced Nephropathy. The
Journal of Urology, Vol. 178, No.6, (December 2007), pp. 2277-2283, ISSN 0022-5347
Toprak, O., Cirit, M., Yesil, M., Bayata, S., Tanrisev, M., Varol, U., Ersoy, R., & Esi, E. (2007).
Impact of Diabetic and Pre-Diabetic State on Development of Contrast-Induced
Nephropathy in Patients with Chronic Kidney Disease. Nephrology Dialysis
Transplantation, Vol.22, No.3, (March 2007), pp. 819-826, ISSN 0931-0509
Toprak, O., Cirit, M., Yesil, M., Byrne DW, Postaci, N., Bayata, S., Majchrzak, KM., & Esi, E.
(2006). Metabolic Syndrome as a Risk Factor for Contrast-Induced Nephropathy in
Non-Diabetic Elderly Patients with Renal Impairment. Kidney and Blood Pressure
Research, Vol.29, No.1, (June 2006), pp. 2-9, ISSN 1420-4096
202 Advances in the Diagnosis of Coronary Atherosclerosis

Toprak, O., Cirit, M., Esi, E., Postaci, N., Yesil, M., & Bayata, S. (2007). Hyperuricemia as a
Risk Factor for Contrast-Induced Nephropathy in Patients with Chronic Kidney
Disease. Catheterization and Cardiovascular Interventions, Vol.67, No.2, (February
2006), pp. 227-235, ISSN 1522-1946
Toprak, O., & Cirit, M. (2006). Risk Factors and Therapy Strategies for Contrast-Induced
Nephropathy. Renal Failure, Vol. 28, No. 5, (January 2006), pp. 365-381, ISSN 0886-022X
Toprak, O., & Cirit, M. (2005). Investigating the Volume Status Before Contrast
Nephropathy Studies. Nephrology Dialysis Transplantation, Vol.20, No.2, (February
2005), pp. 464, ISSN 0931-0509
Toprak, O., Cirit, M., Bayata, S., Yesil, M., & Aslan, SL. (2003). The Effect of Pre-procedural
Captopril on Contrast-Induced Nephropathy in Patients who Underwent Coronary
Angiography. Anadolu Kardiyoloji Dergisi, Vol. 3, No.2, (June 2003), pp. 98-103, ISSN
1302-8723
Toprak, O., Cirit, M., Bayata, S., Aslan, SL., Sarioglu, F., & Cetinkaya, GS. (2003). Is There
Any Relationship Between Left Ventricul Ejection Fraction and Contrast Induced
Nephropathy? Türkiye Klinikleri Journal of Medical Sciences, Vol 23, No. 2, (March
2003), pp. 104-107 , ISSN 1300-0292
Vogt, B., Ferrari, P., Schonholzer, C., Marti, HP., Mohaupt, M., Wiederkehr, M., Cereghetti,
C., Serra, A., Huynh-Do, U., Uehlinger, D., & Frey, FJ. (2001). Prophylactic
Hemodialysis after Radiocontrast Media in Patients with Renal Insufficiency is
Potentially Harmful. The American Journal of Medicine, Vol.111, No.9, (December
2001), pp.692-698, ISSN 0002-9243
Wang, A., Holcslaw, T., Bashore, TM., Freed, MI., Miller, D., Rudnick, MR., Szerlip, H.,
Thames, MD., Davidson, CJ., Shusterman, N., & Schwab, SJ. (2000). Exacerbation of
Radiocontrast Nephrotoxicity by Endothelin Receptor Antagonism. Kidney
International, Vol.57, No.4, (April 2000), pp.1675-1680, ISSN 0085-2538
Weber-Mzell, D., Kotanko, P., Schumacher, M., Klein, W., & Skrabal, F. (2002). Coronary
Anatomy Predicts Presence or Absence of Renal Artery Stenosis. A Prospective
Study in Patients Undergoing Cardiac Catheterization for Suspected Coronary
Artery Disease. European Heart Journal, Vol.23, No.21, (November 2002), pp.1684–
1691, ISSN 0195-668x
Weinrauch, LA., Healy, RW., Leland, OS Jr., Goldstein, HH., Kassissieh, SD., Libertino, JA.,
Takacs, FJ., & D'Elia, JA. (1977). Coronary Angiography and Acute Renal Failure in
Diabetic Azotemic Nephropathy. Annals of Internal Medicine, Vol.86, No.1, (January
1977), pp.56-59, ISSN 0003-4819
Weisberg, LS., Kurnik, PB., & Kurnik, BR. (1994). Risk of Radiocontrast Nephropathy in
Patients with and without Diabetes Mellitus. Kidney International, Vol.45, No.1,
(January 1994), pp.259-265,ISSN 0085-2538
Yang, DW., Jia, RH., Yang, DP., Ding, GH., & Huang, CX. (2004). Dietary
Hypercholesterolemia Aggravates Contrast Media-Induced Nephropathy. Chinese
Medical Journal, Vol. 117, No.4 (April 2004), pp.542–546, ISSN 0366-6999.
Zhang, L., Zhang, L., Lu, Y., Wu, B., Zhang, S., Jiang, H., Ge, J., & Chen, H. (2011). Efficacy of
Statin Pretreatment for the Prevention of Contrast-Induced Nephropathy: A Meta-
Analysis of Randomised Controlled Trials. International Journal of Clinical
Practice,Vol.65, No.5, (May 2011), pp.624-30, ISSN 1368-5031
11

Coronary Angiography in Patients


with Chronic Kidney Disease
Luís Henrique Wolff Gowdak and
José Jayme Galvão de Lima
Heart Institute (InCor), University of
São Paulo Medical School
Brazil

1. Introduction
Cardiovascular disease, mostly coronary artery disease (CAD), remains an important cause
of morbidity and mortality in patients with any degree of renal dysfunction. Presently,
according to the United States Renal Data System, the size of the population with chronic
kidney disease (CKD) stage V reached a new high in 2008, with almost 550,000 patients
under treatment (U.S. Renal Data System, 2010). Sadly, it is expected that half of them will
die from cardiovascular disease; moreover, it is noteworthy that even patients with less
advanced stages of CKD are at greater risk of death from CAD than of reaching the final
stages of renal failure that would eventually require renal replacement therapy (Sarnak et
al., 2003). In these patients, about 50% to 60% of all cardiovascular deaths are due to acute
myocardial infarction, sudden death, or ischemic heart disease (Herzog et al., 2008).
The interplay between CKD and CAD is a very complex one and can only be partly
explained by the fact that patients with CKD share many of the so-called traditional risk
factors also linked to CAD, such as long-standing diabetes, hypertension, low levels of
HDL-cholesterol, and hypertriglyceridemia (McCullough, 2002a). Additionally, a pro-
inflammatory state and high oxidative stress levels are usually seen in patients with
advanced CKD, which may contribute to accelerated atherosclerosis, plaque instability,
acute coronary syndromes, and myocardial fibrosis (Yerkey et al., 2004). The result of this
clustering of traditional and nontraditional risk factors may explain the epidemiological
observation that patients with CKD stage V have more than a 10-fold increased risk of death
from CAD than a patient with 5 Framingham risk scores (McCullough, 2002b).
The prevalence of CAD in CKD patients undergoing dialysis varies with such factors as age,
diabetic status, time of follow-up, and the diligence with which CAD is investigated. In a
group of 4,024 new dialysis patients, including 44% with diabetes, 32% had a history of
CAD (Foley, 2003), which is conceivably more than 3 times the figure corresponding to CAD
in the general population (Levey & Eknoyan, 1999). The prevalence of angiographically
confirmed CAD, on the other hand, defined as luminal reduction >50%, ranges from 24% in
low-risk patients to 85% in older, diabetic patients (Goldsmith & Covic, 2001). Nevertheless,
most of these data originated from studies in which coronary angiography was performed
only in patients with clinical evidence and/or noninvasive studies suggestive of CAD,
204 Advances in the Diagnosis of Coronary Atherosclerosis

leaving out patients in whom asymptomatic, undiagnosed, significant CAD might be


already present; therefore, the actual prevalence of CAD might even be higher.
Adding to the importance of CAD in patients with CKD, observational data also show that
CAD remains a significant clinical problem even if a patient undergoes successful kidney
transplantation (Aakhus et al., 2004). The annual risk of CVD death is 3.5% to 5% in renal
transplant recipients, which is 50-fold higher than that in the general population (Ojo, 2006).
Acute coronary syndrome is highly prevalent in the early posttransplant period, and on
average, CVD mortality accounts for 30% and 75% of early and late posttransplant deaths,
respectively.
Therefore, coronary angiography is a valuable tool, very often used in patients with CKD
both in the acute as well as in the chronic setting of CAD, for the purpose of either the
diagnosis of, or the treatment of CAD by means of percutaneous coronary intervention. The
major challenges with coronary angiography relate to when is it appropriate to perform it as
part of the cardiovascular assessment in patients with CKD and what are the risks
associated with the procedure. The ultimate management of CAD, should it be found after
the diagnostic procedure, is beyond the scope of this chapter.

2. Screening for coronary artery disease in patients with CKD


Based on the above considerations, screening for the presence and severity of CAD in
patients with CKD, and especially in those being considered for renal transplantation, is a
fundamental step during routine cardiovascular risk assessment; in the setting of kidney
transplantation, for instance, numerous studies indicate that not only are the majority of
serious cardiovascular events in renal transplant recipients related to CAD but also these
events tend to occur during the first few months following transplant (Kahwaji et al., 2011).
The last observation strongly suggests that relevant CAD could have been missed before
patients were actually included on renal transplant waiting lists, although they had been
screened for it during routine pretransplant workups according to current guidelines.
According to the American Society of Transplantation recommendations (Kasiske et al.,
2001), patients with diabetes, a prior history of ischemic heart disease or an abnormal ECG,
or age 50 years should be considered at high risk for CAD and referred for a cardiac stress
test, and only in those with a positive stress test, for coronary angiography.
This strategy, however, is under dispute for the now recognized reduced sensitivity and
specificity of noninvasive testing in renal patients compared with that in the general
population (Marwick et al., 1990; Schmidt et al., 2001; Welsh et al., 2011), a finding that
prompted many investigators to recommend direct diagnostic coronary angiography in
high-risk patients. In agreement with the last view, in a previous study, our group showed
that significant CAD, as defined by coronary angiography, was the best predictor of
cardiovascular events compared with radionuclide myocardial perfusion study or
dobutamine-atropine stress echocardiography in renal transplant candidates (De Lima et al.,
2003), even taking into account that coronary angiography is an expensive, invasive
procedure, not free from complications. However, in the same investigation, we also found
that the prevalence of significant CAD was less than 50%, suggesting a need for better
screening mechanisms that could identify high-risk individuals in whom coronary
angiography could be avoided without incurring an increased risk of future events.
Coronary Angiography in Patients with Chronic Kidney Disease 205

2.1 The challenge of referring patients with CKD for coronary angiography
Tables 1 and 2 show the most recent guidelines for coronary angiography to establish the
diagnosis in patients with suspected angina and for risk stratification in patients with chronic
stable angina, respectively (Fraker Jr et al., 2007).
Invasive coronary angiography has received a Class I indication for patients with known or
possible angina pectoris who have survived sudden cardiac death. For risk stratification in
patients with chronic stable angina, coronary angiography should be performed in: a)
patients with disabling (Canadian Cardiovascular Society classes III and IV) stable angina
despite medical therapy; b) patients with high-risk criteria on noninvasive testing; c)
patients with angina and symptoms and signs of heart failure; d) patients with clinical
characteristics indicating a high likelihood of severe CAD.

Coronary angiography is
indicated to establish the Level of
Comment for Patients with CKD
diagnosis of CAD in patients with Evidence
suspected angina in those with…
Known or possible angina pectoris
Survival rates much lower than rates
who have survived sudden cardiac B
for the general population
death

Table 1. Class I indication of the AHA/ACC Guidelines for coronary angiography to


establish a diagnosis in patients with suspected angina in the general population. Modified
from Fraker Jr, et al. (2007).

Coronary angiography is
indicated for risk stratification in Level of
Comment for Patients with CKD
patients with chronic CAD in Evidence
those with…
Disabling chronic stable angina Uncommon clinical presentation for
B
despite medical therapy patients with CKD
High-risk criteria on noninvasive
Lower sensitivity than that in the
testing regardless of angina B
general population
severity
Angina who have survived sudden
Survival rates much lower than those
cardiac death or serious ventricular B
in the general population
arrhythmia
May be confused with hypertensive
Angina and symptoms and signs of cardiomyopathy, fluid overload,
C
congestive heart failure
subdialysis
Clinical characteristics that indicate Lower specificity than that in the
C
a high likelihood of severe CAD general population

Table 2. Class I indications of the AHA/ACC Guidelines for coronary angiography for risk
stratification in patients with chronic stable angina. Modified from Fraker Jr, et al. (2007).
206 Advances in the Diagnosis of Coronary Atherosclerosis

Although these guidelines have been extensively tested and validated in the general
population, for patients with CKD the data supporting that approach is less robust. The
main pitfalls of applying the same criteria recommended for patients with normal renal
function to patients with CKD are briefly described below.

2.1.1 Clinical presentation


Although angina pectoris is usually a reliable indicator of the presence of CAD in the
general population, it is not so among patients with CKD, particularly in the more advanced
stages of renal failure. For instance, chest pain is commonly observed in the dialysis
population; nevertheless, its characteristics make the discomfort often described as
“atypical,” and the pain does not correlate with the presence of significant CAD. In fact,
fewer than 50% of dialysis patients with acute myocardial infarction present with chest pain,
which likely explains why myocardial infarction is infrequently suspected on admission
(Herzog et al., 2007). Reasons for the different presentations of ischemia are not well
understood but may be related to associated diabetic or uremic neuropathy. Diagnosis of
ischemia is further complicated by the fact that other symptoms of ischemia, such as
dyspnea on exertion, fatigue, and hypotension, are common in dialysis patients and may be
attributed to dialysis-related factors, such anemia, volume overload, acidosis, non-
compliance with fluid intake or dialysis or to ultra-filtration during dialysis (Sosnov et al.,
2006). Furthermore, silent ischemia occurs frequently in patients with CKD.
We previously found that, in a group of high-risk renal transplant candidates who
underwent coronary angiography per protocol, regardless of symptoms, the prevalence of
significant CAD was 40% in patients with, and 58% in patients without, angina (De Lima et
al., 2003). Disabling symptoms, even in the presence of significant and extensive CAD, are
quite uncommon in patients with CKD.

2.1.2 Noninvasive testing


2.1.2.1 Resting and exercise electrocardiogram
The interpretation of resting and exercise electrocardiograms for the diagnosis of CAD in
patients on dialysis is often hampered by the presence of concurrent left ventricular
hypertrophy, anemia, and electrolyte disturbances that may cause electrocardiographic
changes resembling those of ischemia (Sharma et al., 2007). Also, most patients with CKD
have poor physical stamina, limiting the utility of exercise electrocardiography as a
diagnostic tool. The overall specificity and sensitivity of both tests for the diagnosis of CAD
are lower (<50%) in patients with CKD than in the general population.
2.1.2.2 Myocardial perfusion assessment
Dipyridamole radionuclide stress testing with single-photon emission tomography (SPECT) has
been widely used as a screening test for CAD in patients with a variety of clinical conditions.
The reported specificity and sensitivity of the test are close to 70% and 80%, respectively.
Among patients with CKD, however, the results are highly variable with sensitivity often <60%
compared to that of coronary angiography (Holley et al., 1991; Sharma et al., 2007). The reasons
for the unexpected low sensitivity and great variability of cardiac scintigraphy in patients with
CKD are unclear but may reflect the increased levels of circulating adenosine, reduced coronary
flow reserve, or left ventricular hypertrophy. Combined dipyridamole-exercise thallium
imaging reportedly increases the sensitivity and specificity of the test.
Dobutamine-atropine stress echocardiography has been associated with better and more
consistent results for detecting significant (>70%) coronary stenosis in patients with CKD,
Coronary Angiography in Patients with Chronic Kidney Disease 207

with a sensitivity and negative predictive value between 70% and 80% (Herzog et al., 1999;
Ferreira et al., 2007). However, the utility of this test is limited, because dobutamine often
increases blood pressure, causing interruption of the test in about 15% of patients.
Until a few years ago, magnetic resonance imaging (MRI) had been used for assessment of
myocardial function and detection of myocardial ischemia and scar tissue in patients with
CKD (Andrade et al., 2009). Myocardial stress perfusion studies performed with MRI
adequately detected CAD, and there was a good correlation between these results and those
from coronary angiography and radionuclide studies (Andrade et al., 2004). Unfortunately,
further development of this imaging modality was halted, because of the description of a
serious complication related to the use of gadolinium-based contrast media in patients with
CKD, called nephrogenic systemic fibrosis (Grobner, 2006). Thus, starting in 2007, the
United States Food and Drug Administration issued a boxed warning about this
progressive, debilitating and, occasionally fatal condition recommending that the use of
gadolinium-based contrast agents should be avoided in patients with acute or chronic renal
insufficiency with a glomerular filtration rate < 30mL/min/1.73m2.
2.1.2.3 Coronary angiotomography
Multidetector-row computed tomography (MDCT) has been recently applied to the
detection of CAD in patients with ESRD (Rosário et al., 2010). This method allows not only
the detection of coronary calcifications but also the detailed visualization of the arterial wall
and lumen, and therefore, may become an alternative to coronary angiography. Experience
with this method in patients with CKD is still very limited.
2.1.2.4 Clinical implications
Thus, referring patients with CKD for coronary angiography is not an easy task (De Lima et
al., 2010; Lentine et al., 2010). One could be very “conservative,” only referring patients with
overt, unequivocal symptoms of angina or extensive myocardial ischemia on noninvasive
testing to undergo coronary angiography.
This approach would most likely yield a prohibitively large number of “false negatives,”
meaning that many patients with significant CAD will be deemed free of disease. It is quite
easy to see the clinical implications of such an approach, especially for renal transplant
candidates. Alternatively, one could be very “aggressive” and refer all patients with CKD
stages IV or V for coronary angiography. We would be able to diagnose every single patient
with CAD in adopting this strategy. The downside is that coronary angiography is a
somewhat risky, costly procedure and as many as 50% to 60% of all patients will have no
significant CAD at all and, therefore, will not be further referred for coronary interventions
(percutaneous or surgical). Finally, patients still not on renal replacement therapy might not
only not benefit from this strategy but might also have worsening of their residual renal
function because of the contrast-induced nephropathy.
As a matter of fact, it is still unclear whether the benefits outweigh the harms of routine
screening and prophylactic revascularization to prevent coronary artery disease (CAD) in
asymptomatic kidney transplant candidates. To help clarify that issue, an ongoing
randomized clinical trial is being performed to study the effect of CAD screening on major
adverse cardiac events (Kasiske et al., 2011).

3. In whom should coronary angiography be performed?


The same indications presented in Tables 1 and 2 should be applied to patients with CKD.
The greatest challenge facing the clinical cardiologist called upon to assess a patient with
208 Advances in the Diagnosis of Coronary Atherosclerosis

CKD regarding the presence and severity of CAD is, based on the previous considerations,
which additional patients should be referred for coronary angiography, especially those
asymptomatic patients being assessed before renal transplantation.
In a previous study, we sought to determine the clinical predictors more closely related to
CAD in 301 renal transplant candidates treated by hemodialysis (Gowdak et al, 2007). CAD
(> 70% stenosis) was found in 45% of patients, and the clinical variables significantly
associated with CAD were diabetes, peripheral vascular disease, and previous myocardial
infarction. More important, the prevalence of CAD increased with the number of clinical
predictors from 26% (none) to 100% (all present), while the incidence of events increased 2-,
4-, and 6-fold in those with diabetes, peripheral vascular disease, or previous myocardial
infarction, respectively (p < .0001). This approach would allow reducing the prevalence of
unnecessary angiography from 55% (when all patients undergo angiography) to 26%.
However, we believe that missing the diagnosis of CAD in one-fourth of patients is still not
totally satisfactory.
Another approach should be to restrict the use of coronary angiography as a predictive tool
of cardiovascular events in patients with CKD in which this strategy is powerful enough to
discern between patients at high- and low-risk, comparatively to either clinical stratification
alone or to noninvasive testing. We compared the efficacy of clinical stratification,
noninvasive testing, and coronary angiography in predicting cardiovascular events in
patients with CKD being considered as potential renal transplant candidates (De Lima et al.,
2006; Gowdak et al., 2008). We were able to show that, for patients considered at low-risk for
CAD (age < 50 years, with no diabetes, and no history of cardiovascular disease), additional
noninvasive or invasive testing, added no capability in predicting cardiovascular events
beyond that given by clinical stratification. On the other hand, for patients at high-risk for
CAD, defined by the presence of any 2 risk factors combined (age  50 years or diabetes or
cardiovascular disease), noninvasive testing failed to identify patients at higher risk for
cardiovascular events; indeed, the finding of significant CAD by angiography (luminal
stenosis  70%) was the strongest predictor of events. For patients at intermediate-risk for
CAD, defined by the presence of any single risk factor alone, noninvasive testing was a good
tool in identifying patients at higher risk of events, should an abnormal myocardial
perfusion scan be found. Figure 1 shows the proposed algorithm for risk stratification of
CAD in renal transplant candidates.

4. Contrast-induced Acute Kidney Injury


Very often, the highlighted importance of coronary angiography for the diagnosis and/or
management of CAD in patients with CKD is offset by the possibility of contrast-induced
acute kidney injury (AKI) that may follow the use of any iodinated contrast media.
Contrast-induced AKI is a common and potentially serious complication after the
administration of contrast media in patients at risk for acute renal injury (McCullough,
2008). Unfortunately, the most important risk factor for contrast-induced AKI is an already
compromised baseline renal function. The knowledge that not only contrast-induced AKI
adversely affects the short- and long-term prognosis but also implies greater health costs
prompted cardiologists to be particularly careful whenever coronary angiography is
recommended to patients with any degree of CKD.
Many definitions currently exist in practice regarding contrast-induced AKI; it can be
simply defined as an increase in serum creatinine (SCr) occurring within the first 24h after
contrast exposure and peaking up to 5 days afterwards. The rise in Scr can be expressed
Coronary Angiography in Patients with Chronic Kidney Disease 209

either in absolute terms (0.5 to 1.0mg/dL) or as a proportional rise in SCr of 25% to 50%
above the baseline value (Gleeson & Bulugahapitiya, 2004).

RENAL TRANSPLANT CANDIDATES

Clinical stratification following the 1. Age  50 years


American Society of Transplantation 2. Diabetes
Guidelines 3. History of cardiovascular disease

NUMBER OF RISK FACTORS

ZERO ONE TWO THREE

LOW RISK INTERMEDIATE HIGH RISK


RISK

NON-INVASIVE
TESTING

NEGATIVE POSITIVE

CORONARY ANGIOGRAPHY

No further NEGATIVE POSITIVE


testing

Treat according to the current


guidelines

Fig. 1. Proposed algorithm for coronary angiography in patients with CKD awaiting renal
transplantation.
210 Advances in the Diagnosis of Coronary Atherosclerosis

The increase in SCr seen in patients with contrast-induced AKI is associated with a
significant increase in the risk of death. In a large retrospective study comprising over 16,000
patients undergoing procedures, those who developed contrast-induced AKI had a 5.5-fold
increased risk of death during hospitalization (Levy et al., 1996). Another study with more
than 7,000 patients confirmed the short-term worse prognosis in patients with AKI but
extended the greater risk of death up to 5 years after the procedure: the mortality rates at 1
year after development of contrast-induced AKI (12.1%) and at 5 years (44.6%) were higher
compared with rates of 3.7% and 14.5%, respectively, in patients who did not develop
contrast-induced AKI, indicating that the increased risk of death persisted in the long term
(Rihal et al., 2002).
Besides the increased mortality, contrast-induced AKI also poses a high morbidity risk. Its
occurrence has been linked to late cardiovascular events after percutaneous coronary
intervention (such as myocardial infarction), bleeding requiring transfusion, and vascular
complications. As a consequence of more serious adverse events, the in-hospital stay is
longer with an estimated US$10,000 to US$12,000 added to the final bill during the first year
(Subramanian et al., 2007).

4.1 Pathophysiology of contrast-induced Acute Kidney Injury


Centrally to the pathophysiology of contrast-induced AKI is the presence of a reduced
nephron mass, a hallmark of chronic kidney disease. It has already been demonstrated that
in patients with CKD, defined by an estimated glomerular filtration rate <
60mL/min/1.73m2, there is a considerable loss of nephron units, making the residual renal
function prone to further declines with renal insults (Figure 2). When serum creatinine or
eGFR is unavailable, then a survey may be used to identify patients at higher risk than the
general population for contrast-induced AKI. Iodinated contrast, after causing a brief
(minutes) period of vasodilation, causes sustained (hours to days) intrarenal
vasoconstriction and ischemic injury. The ischemic injury sets off a cascade of events largely
driven by oxidative injury causing death of renal tubular cells. If a sufficient mass of
nephron units is affected, then a recognizable rise in serum creatinine will occur. Any
superimposed insult, such as sustained hypotension in the catheterization laboratory, micro-
showers of atheroembolic material from catheter exchanges or the use of intraaortic balloon
counterpulsation, or a bleeding complication can amplify the injury processes occurring in
the kidney (McCullough et al., 2006a).
Renal function must be assessed before administration of contrast medium so appropriate
caution may be exercised to avoid additional risks. The National Kidney Foundation Kidney
Disease Outcome Quality Initiative recommends that clinicians use an eGFR calculated from
the SCr as an index of renal function rather than using SCr in stable patients. In such a
situation when an eGFR cannot be obtained prior to the procedure, clinicians should inquire
about risk markers for AKI after iodinated contrast (Table 3).
The presence of multiple contrast-induced AKI risk factors in the same patient or high-risk
clinical scenarios can create a very high risk (50%) for contrast-induced AKI and (15%)
acute renal failure requiring dialysis after contrast exposure.
In the setting of emergency procedures, where the benefit of very early imaging outweighs
the risk of waiting, the procedure can be performed without knowledge of serum creatinine
or eGFR.
Coronary Angiography in Patients with Chronic Kidney Disease 211

Risk markers for contrast-induced acute kidney injury

previous renal disease diabetes mellitus

prior renal surgery hypertension

proteinuria gout

use of nephrotoxic drugs, such as


nonsteroidal anti-inflammatory agents

Table 3. A brief 7-item survey to identify patients at higher risk for AKI.

Fig. 2. Postulated pathophysiology of contrast-induced AKI (McCullough, 2008).

4.2 Contrast medium use


In patients at increased risk for contrast-induced AKI undergoing intraarterial
administration of contrast, ionic high-osmolality agents pose a greater risk for contrast-
induced AKI than low-osmolality agents (Barrett & Carlisle, 1993). Red blood cell
deformation, systemic vasodilation, intrarenal vasoconstriction, as well as direct renal
tubular toxicity are all more common in contrast agents with osmolality greater than that of
blood.
Current evidence suggests that for intraarterial administration in high-risk patients with
CKD, particularly those with diabetes mellitus, nonionic, iso-osmolar contrast is associated
212 Advances in the Diagnosis of Coronary Atherosclerosis

with the lowest risk of contrast-induced AKI. Iodixanol (an iso-osmolar contrast media) has
been shown to have the lowest risk for contrast-induced AKI in patients with CKD and
diabetes mellitus compared with low-osmolar contrast media (McCullough et al. 2006b). The
American College of Cardiology/American Heart Association guidelines for the management of
acute coronary syndromes in patients with CKD listed the use of iso-osmolar contrast media
as a class I, Level of Evidence: A recommendation. The National Kidney Foundation Kidney
Disease Outcome Quality Initiative guidelines have also recommended use of iso-osmolar
contrast media in renal dialysis patients to minimize the chances of volume overload and
complications before the next dialysis session.
Higher contrast volumes (100 mL) are associated with higher rates of contrast-induced
AKI in patients at risk. However, even small (30 mL) volumes of iodinated contrast in
very high-risk patients can cause contrast-induced AKI and acute renal failure requiring
dialysis, suggesting the absence of a threshold effect (Manske et al., 1990). As a general
rule, the volume of contrast received should not exceed twice the baseline level of eGFR in
mL.
Finally, intraarterial administration of iodinated contrast appears to pose a greater risk of
contrast-induced AKI above that with intravenous administration.

4.3 Strategies to further reduce the risk of contrast-induced Acute Kidney Injury
4.3.1 Volume expansion
Adequate intravenous volume expansion with isotonic crystalloid (1.0–1.5 mL/kg/h) for 3–
12 h before the procedure and continued for 6–24 h afterwards can lessen the probability of
contrast-induced AKI in patients at risk (Mueller et al., 2002). Achieving a good urine output
(150 mL/h) in the 6h after the procedure has been associated with reduced rates of AKI in
one study. The data on oral as opposed to intravenous volume expansion as a contrast-
induced AKI prevention measure are insufficient.

4.3.2 Pharmacologic agents


No adjunctive medical or mechanical treatment has been proven to be efficacious reducing
the risk of AKI after exposure to iodinated contrast. With iodinated contrast, the
pharmacologic agents tested in small trials that deserve further evaluation include the
antioxidants ascorbic acid and N-acetylcysteine (NAC), aminophylline/theophylline,
statins, and prostaglandin E1.
4.3.2.1 Ascorbic acid
Only ascorbic acid has been tested in a multicenter, blinded, placebo-controlled trial (n=231)
and been shown to reduce rates of contrast-induced AKI. The dose of ascorbic acid (vitamin
C over the counter) used in this trial was 3g orally the night before and 2g orally twice a day
after the procedure (Spargias et al., 2004).
4.3.2.2 N-acetylcysteine
Although popular, NAC has not been consistently shown to be effective. Nine meta-
analyses have been published, all documenting the significant heterogeneity between
studies and pooled odds ratios for NAC approaching unity (Stacul et al., 2006). Importantly,
only in those trials where NAC reduced SCr below baseline values because of decreased
skeletal muscle production did renal injury rates appear to be reduced.
Coronary Angiography in Patients with Chronic Kidney Disease 213

However, NAC as an antioxidant has been shown to lower rates of AKI and mortality after
primary PCI in 1 trial. The recently published REMEDIAL (Renal Insufficiency Following
Contrast Media Administration) trial suggested that the use of volume supplementation
with sodium bicarbonate together with NAC was more effective than NAC alone in
reducing the risk of AKI (Briguori et al., 2007; Brown et al., 2009). Dosing of NAC has varied
in the trials; however, the most successful approach has been with 1,200 mg orally twice a
day on the day before and after the procedure.
4.3.2.3 Other agents
Fenoldopam, dopamine, calcium-channel blockers, atrial natriuretic peptide, and L-arginine
have not been shown to be effective in the prevention of contrast-induced AKI. Furosemide,
mannitol, and an endothelin receptor antagonist are potentially detrimental (Stacul et al.,
2006).
In general, cardiovascular patients undergoing procedures with iodinated contrast have
either high risk for atherosclerosis or have the anatomic presence of disease. Therefore, the
vast majority of patients should be on statin therapy with a common low-density lipoprotein
cholesterol target of <70 mg/dL. Several studies have demonstrated that patients continuing
on statins during cardiovascular procedures including PCI and coronary artery bypass
grafting have lower rates of AKI (Khanal et al., 2005). All small, randomized trials published
to date support this concept as well. Preservation of endothelial function at the level of the
glomerulus and reductions in systemic inflammatory factors are postulated mechanisms by
which statins may have renoprotective effects.
4.3.2.4 Dialysis and hemofiltration
Prophylactic hemodialysis or hemofiltration has not been validated as an effective strategy,
even when carried out within 1 h or simultaneously with contrast administration.
Hemofiltration, however, performed 6 h before and 12 to 18 h after contrast deserves
consideration, given reports of reduced mortality and need for hemodialysis in the
postprocedure period in very high-risk patients (Marenzi et al., 2003).
4.3.2.5 Withdrawal of additional nephrotoxic drugs
Despite the lack of more robust data in this area, it is a reasonable practice to withhold
nonsteroidal anti-inflammatory drugs, calcineurin inhibitors, high-dose loop diuretics,
aminoglycosides, and other nephrotoxic agents if possible for several days before contrast
exposure (McCullough, 2008). It is routine practice to withhold metformin before all contrast
procedures, not because metformin itself is nephrotoxic but because in the setting of AKI if
metformin is continued, lactic acidosis can develop leading to systemic complications and
death. As a general rule, metformin should not be restarted until the clinician is confident
that the patient has not developed AKI.

4.4 Management of patients receiving iodinated contrast media


An integrated proposed algorithm for the management of patients receiving iodinated
contrast media is presented in Figure 3. Since there are no approved pharmaceutical agents
for the prevention of this complication, the practitioner should be cautious with the use of
any of the drugs suggested. We also recommend that all patients at risk for contrast-induced
AKI should have follow-up serum creatinine and electrolyte monitoring daily while in the
hospital, and then at 48 to 96h after discharge.
214 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 3. Proposed algorithm for management of contrast-induced AKI (McCullough, 2008).

5. Conclusion
Patients with chronic kidney disease are at high-risk for CAD and, therefore, coronary
angiography is very often needed to establish the diagnosis and severity of CAD. The major
challenges in this regard are how to select those patients more likely to have significant
CAD, in the absence of typical symptoms suggestive of CAD or myocardial perfusion
defects disclosed by noninvasive testing. Moreover, the possibility of contrast-induced acute
kidney injury should signal the need for a careful and more judicious decision-making
approach by the cardiologist, working in tandem with the nephrologist, when trying to
establish the risk-benefit ratio for each patient being considered for invasive assessment.
Having decided that an invasive procedure is clearly needed, all preventive and therapeutic
measurements should be applied to minimize the risk of worsening residual renal function.

6. References
Aakhus S, Dahl K & Widerøe TE (2004). Cardiovascular disease in stable renal transplant
patients in Norway: morbidity and mortality during a 5-yr follow-up. Clinical
Transplantation, Vol.18, No.5, pp.596-604.
Andrade JM, Rochitte CE, Gowdak LH, Paula FJ, Giorgi MCP, Parga JR, Avila LF, Ramires
JAF & Lima JJG (2004). Evaluation of myocardial ischemia by magnetic resonance
myocardial stress perfusion in renal transplant candidates: Comparison with
SPECT and coronary angiography. Journal of the American College of Cardiology,
Vol.43, No.5 (Supplement A), pp.353A-353A.
Coronary Angiography in Patients with Chronic Kidney Disease 215

Andrade JM, Gowdak LH, Giorgi MC, de Paula FJ, Kalil-Filho R, de Lima JJ & Rochitte CE
(2009). Cardiac MRI for detection of unrecognized myocardial infarction in patients
with end-stage renal disease: comparison with ECG and scintigraphy. American
Journal of Roentgenolology, Vol.193, No.1, pp.W25-32.
Barrett BJ & Carlisle EJ (1993). Metaanalysis of the relative nephrotoxicity of high- and low-
osmolality iodinated contrast media. Radiology, Vol.188, No.1, pp.171-178.
Briguori C, Airoldi F, D'Andrea D, Bonizzoni E, Morici N, Focaccio A, Michev I, Montorfano
M, Carlino M, Cosgrave J, Ricciardelli B & Colombo A (2007). Renal insufficiency
following contrast media administration trial (REMEDIAL): a randomized
comparison of 3 preventive strategies. Circulation, Vol.115, No.10, pp.1211–1217.
Brown JR, Block CA, Malenka DJ, O'Connor GT, Schoolwerth AC & Thompson CA (2009).
Sodium bicarbonate plus N-acetylcysteine prophylaxis: a meta-analysis. Journal of
the American College of Cardiology Cardiovascular Interventions, Vol.2, No.11, pp.1116-
1124.
De Lima JJ, Sabbaga E, Vieira ML, de Paula FJ, Ianhez LE, Krieger EM & Ramires JA (2003).
Coronary angiography is the best predictor of events in renal transplant candidates
compared with noninvasive testing. Hypertension, Vol. 42, No.3, pp.263-268.
De Lima JJG, Paula FJ, Martinez Filho E, Meneghetti C, Cesar LAM, Ramires JAF, Krieger
EM & Gowdak LHW (2006). Predicting cardiovascular events in patients with end-
stage renal disease on the waiting list for renal transplantation - to invade or not to
invade? European Heart Journal, Vol.27, Supplement 1, pp.10-10.
De Lima JJ, Wolff Gowdak LH, de Paula FJ, Ianhez LE, Franchini Ramires JA & Krieger EM
(2010). Validation of a strategy to diagnose coronary artery disease and predict
cardiac events in high-risk renal transplant candidates. Coronary Artery Disease,
Vol.21, No.3, pp.164-167.
Ferreira PA, de Lima VC, Campos Filho O, Gil MA, Cordovil A, Machado CV, Pestana JO &
Carvalho AC (2007). Feasibility, safety and accuracy of dobutamine/atropine stress
echocardiography for the detection of coronary artery disease in renal transplant
candidates. Arquivos Brasileiros de Cardiologia, Vol.88, No.1, pp.45-51.
Foley RN (2003). Clinical epidemiology of cardiac disease in dialysis patients: left
ventricular hypertrophy, ischemic heart disease, and cardiac failure. Seminars in
Dialysis, Vol.16, No.2, pp.111-117.
Fraker Jr TD, Fihn SD & Writing on behalf of the 2002 Chronic Stable Angina Writing
Committee (2007). 2007 chronic angina focused update of the ACC/AHA 2002
guidelines for the management of patients with chronic stable angina: a report of
the American College of Cardiology/American Heart Association Task Force on
Practice Guidelines Writing Group to develop the focused update of the 2002
guidelines for the management of patients with chronic stable angina. Journal of the
American College of Cardiology, Vol.50, No.23, pp.2264-2274.
Gleeson TG & Bulugahapitiya S (2004). Contrast-induced nephropathy. American Journal of
Roentgenology, Vol.183, No.6, pp.1673-1789.
Goldsmith DJ & Covic A (2001). Coronary artery disease in uremia: Etiology, diagnosis, and
therapy. Kidney International, Vol.60, No. 6, pp.2059-2078.
Gowdak LH, de Paula FJ, César LA, Martinez Filho EE, Ianhez LE, Krieger EM, Ramires JA
& de Lima JJ (2007). Screening for significant coronary artery disease in high-risk
renal transplant candidates. Coronary Artery Disease, Vol.18, No.7, pp.553-558.
216 Advances in the Diagnosis of Coronary Atherosclerosis

Gowdak LHW, Arantes RL, De Paula FJ, Cesar LAM, Ianhez LE, Krieger EM & De Lima JJG
(2008). Reviewing the American Society of Transplantation guidelines for
cardiovascular risk stratification in renal transplant candidates. Circulation, Vol.118,
No.12, pp.E462-E462.
Grobner T (2006). Gadolinium – a specific trigger for the development of nephrogenic
fibrosing dermopathy and nephrogenic systemic fibrosis? Nephrolology Dialysis
Transplantation, Vol.21, No.4, pp.1104-1108.
Herzog CA, Marwick TH, Pheley AM, White CW, Rao VK & Dick CD (1999). Dobutamine
stress echocardiography for the detection of significant coronary artery disease in
renal transplant candidates. American Journal of Kidney Disease, Vol.33, No.6,
pp.1080-1090.
Herzog CA, Littrell K, Arko C, Frederick PD & Blaney M (2007). Clinical characteristics of
dialysis patients with acute myocardial infarction in the United States: a
collaborative project of the United States Renal Data System and the National
Registry of Myocardial Infarction. Circulation, Vol.116, No.13, pp.1465-1472.
Herzog CA, Mangrum JM & Passman R (2008). Sudden cardiac death and dialysis patients.
Seminars in Dialysis, Vol. 21, No.4, pp.300-307.
Holley JL, Fenton RA & Arthur RS (1991). Thallium stress testing does not predict
cardiovascular risk in diabetic patients with end-stage renal disease undergoing
cadaveric renal transplantation. American Journal of Medicine, Vol.90, No.5, pp.563-
570.
Kahwaji J, Bunnapradist S, Hsu JW, Idroos ML & Dudek R (2011). Cause of death with graft
function among renal transplant recipients in an integrated healthcare system.
Transplantation, Vol. 91, No.2, pp.225-230.
Kasiske BL, Cangro CB, Hariharan S, Hricik DE, Kerman RH, Roth D, Rush DN, Vazquez
MA, Weir MR & American Society of Transplantation (2001). The evaluation of
renal transplantation candidates: clinical practice guidelines. American Journal of
Transplantation, Vol. 1, Supplement 2, pp.3-95.
Kasiske BL, Israni AK, Snyder JJ, Camarena A & COST Investigators (2011). Design
considerations and feasibility for a clinical trial to examine coronary screening
before kidney transplantation (COST). American Journal of Kidney Disease, Mar 14,
Epub ahead of print.
Khanal S, Attallah N, Smith DE, Kline-Rogers E, Share D, O'Donnell MJ & Moscucci M
(2005). Statin therapy reduces contrast-induced nephropathy: an analysis of
contemporary percutaneous interventions. American Journal of Medicine, Vol.118,
No.8, pp.843-849.
Lentine KL, Hurst FP, Jindal RM, Villines TC, Kunz JS, Yuan CM, Hauptman PJ & Abbott
KC (2010). Cardiovascular risk assessment among potential kidney transplant
candidates: approaches and controversies. American Journal of Kidney Disease,
Vol.55, No.1, pp.152-167.
Levey AS & Eknoyan G (1999). Cardiovascular disease in chronic renal disease. Nephrology
Dialysis Transplantation,. Vol. 14, No.4, pp.828-833.
Levy EM, Viscoli CM & Horwitz RI (1996). The effect of acute renal failure on mortality. A
cohort analysis. Journal of the American Medical Association, Vol.275, No.19, pp.1489-
1494.
Coronary Angiography in Patients with Chronic Kidney Disease 217

Manske CL, Sprafka JM, Strony JT & Wang Y (1990). Contrast nephropathy in azotemic
diabetic patients undergoing coronary angiography. American Journal of Medicine,
Vol.89, No.5, pp.615-620.
Marenzi G, Marana I, Lauri G, Assanelli E, Grazi M, Campodonico J, Trabattoni D,
Fabbiocchi F, Montorsi P & Bartorelli AL (2003). The prevention of radiocontrast
agent-induced nephropathy by hemofiltration. The New England Journal of Medicine,
Vol.349, No.14, pp.1333-1340.
Marwick TH, Steinmuller DR, Underwood DA, Hobbs RE, Go RT, Swift C & Braun WE
(1990). Ineffectiveness of dipyridamole SPECT thallium imaging as a screening
technique for coronary artery disease in patients with end-stage renal failure.
Transplantation, Vol.49, No.1, pp.100-103.
McCullough PA (2002a). Scope of cardiovascular complications in patients with kidney
disease. Ethnicity and Disease, Vol.12, No.4, pp.S3-44-48.
McCullough PA (2002b). Cardiorenal risk: An important clinical intersection. Reviews in
Cardiovascular Medicine, Vol.3, No.2, pp.71-76.
McCullough PA, Adam A, Becker CR, Davidson C, Lameire N, Stacul F, Tumlin J & CIN
Consensus Working Panel (2006a). Risk prediction of contrast-induced
nephropathy. American Journal of Cardiology, Vol.98, No.6A, pp.27K–36K.
McCullough PA, Bertrand ME, Brinker JA & Stacul F (2006b). A meta-analysis of the renal
safety of isosmolar iodixanol compared with low-osmolar contrast media. Journal of
the American College of Cardiology, Vol.48, No.4, pp.692–699.
McCullough PA (2008). Contrast-induced acute kidney injury. Journal of the American College
of Cardiology, Vol.51, No.15, pp.1419-1428.
Mueller C, Buerkle G, Buettner HJ, Petersen J, Perruchoud AP, Eriksson U, Marsch S &
Roskamm H (2002). Prevention of contrast media-associated nephropathy:
randomized comparison of 2 hydration regimens in 1620 patients undergoing
coronary angioplasty. Archives of Internal Medicine, Vol.162, No.3, pp.329-336.
Ojo AO (2006). Cardiovascular complications after renal transplantation and their
prevention. Transplantation, Vol.82, No.5, pp. 603-611.
Rihal CS, Textor SC, Grill DE, Berger PB, Ting HH, Best PJ, Singh M, Bell MR, Barsness GW,
Mathew V, Garratt KN & Holmes Jr DR (2002). Incidence and prognostic
importance of acute renal failure after percutaneous coronary intervention.
Circulation, Vol.105, No.19, pp.2259-2264.
Rosário MA, Lima JJ, Parga JR, Avila LF, Gowdak LH, Lemos PA & Rochitte CE (2010).
Coronary calcium score as predictor of stenosis and events in pretransplant renal
chronic failure. Arquivos Brasileiros de Cardiologia, Vol.94, No.2, pp.252-260.
Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL, McCullough PA,
Kasiske BL, Kelepouris E, Klag MJ, Parfrey P, Pfeffer M, Raij L, Spinosa DJ &
Wilson PW; American Heart Association Councils on Kidney in Cardiovascular
Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology
and Prevention (2003). Kidney disease as a risk factor for development of
cardiovascular disease: a statement from the American Heart Association Councils
on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical
Cardiology, and Epidemiology and Prevention. Circulation, Vol.108, No.17,
pp.2154-2169.
218 Advances in the Diagnosis of Coronary Atherosclerosis

Sharma R, Pellerin D & Brecker SJ (2007). The detection of myocardial ischemia in end-stage
renal disease. Current Opinion in Investigational Drugs, Vol.8, No.3, pp.232-236.
Schmidt A, Stefenelli T, Schuster E & Mayer G (2001). Informational contribution of
noninvasive screening tests for coronary artery disease in patients on chronic renal
replacement therapy. American Journal of Kidney Disease, Vol.37, No.1, pp.56-63.
Sosnov J, Lessard D, Goldberg RJ, Yarzebski J & Gore JM (2006). Differential symptoms of
acute myocardial infarction in patients with kidney disease: a community-wide
perspective. American Journal of Kidney Disease, Vol.47, No.3, pp.378-384.
Spargias K, Alexopoulos E, Kyrzopoulos S, Iokovis P, Greenwood DC, Manginas A, Voudris
V, Pavlides G, Buller CE, Kremastinos D & Cokkinos DV (2004). Ascorbic acid
prevents contrast-mediated nephropathy in patients with renal dysfunction
undergoing coronary angiography or intervention. Circulation, Vol.110, No.18,
pp.2837-2842.
Stacul F, Adam A, Becker CR, Davidson C, Lameire N, McCullough PA, Tumlin J & CIN
Consensus Working Panel (2006). Strategies to reduce the risk of contrast-induced
nephropathy. American Journal of Cardiology, Vol.98, No.6A, pp.59K–77K.
Subramanian S, Tumlin J, Bapat B & Zyczynski T (2007). Economic burden of contrast-
induced nephropathy: implications for prevention strategies. Journal of Medical
Economics, Vol.10, No.2, pp.119-134.
U.S. Renal Data System (2010). USRDS 2010 Annual Data Report: Atlas of Chronic Kidney
Disease and End-Stage Renal Disease in the United States, National Institutes of
Health, National Institute of Diabetes and Digestive and Kidney Diseases,
Bethesda, MD. Accessed: Apr 26, 2011, Available from: <http://www.usrds.org>
Welsh RC, Cockfield SM, Campbell P, Hervas-Malo M, Gyenes G & Dzavik V (2011).
Cardiovascular assessment of diabetic end-stage renal disease patients before renal
transplantation. Transplantation, Vol.91, No.2, pp.213-218.
Yerkey MW, Kernis SJ, Franklin BA, Sandberg KR & McCullough PA (2004). Renal
dysfunction and acceleration of coronary disease. Heart, Vol. 90, No. 8, pp.961-966.
12

Cardiac Catheterization and Coronary


Angiography in Patients
with Cardiomyopathy
Ali Ghaemian
Mazandaran University of Medical Sciences
Iran

1. Introduction
Cardiomyopathies are a group of heart diseases that influence muscles directly and are not
related to congenital, valvular, and pericardial diseases (Salehifar et al., 2008).
Cardiomyopathies are usually classified on the basis of structural or functional causes. Thus,
dilated cardiomyopathy is characterized by an enlarged ventricular chamber size and
reduced cardiac contractility, hypertrophic cardiomyopathy is characterized by hypertrophy
of the ventricular walls and increased cardiac performance and finally the restrictive form is
characterized by stiff ventricular walls which restrict diastolic filling of the ventricle while
systolic performance is not impaired (Ammash et al., 2000; Dec & Fuster ,1994). The
classification by the American Heart Association Scientific Statement panel divides
cardiomyopathy into primary and secondary causes (Maron et al., 2006).In this classification
primary cardiomyopathies are subdivided into genetic, mixed and acquired groups.

2. Dilated cardiomyopathy
Dilated cardiomyopathy (DCM) is characterized by enlargement of one or both ventricles
accompanied by impaired myocardial systolic function.DCM is the most common
cardiomyopathy. Patients with DCM usually have symptoms and signs of congestive heart
failure and there are many cardiac and systemic causes leading to DCM. However, in about
50 percent or more of the patients with DCM, the etiology can not be identified. Thus, the
patient is called as having an idiopathic DCM (Felker et al., 2000).

2.1 Natural history and prognosis


Because of different etiologies patients have variable presentations. Asymptomatic patients
with left ventricular dysfunction to advanced stages of heart failure are two sides of the
spectrum of presentations of the patients. The annual mortality ranges between 10 and 50
percents (Levy et al., 2006).While survival of some patients is good, some cardiomyopathy
patients have poor prognosis. For example, cardiomyopathy patients with human
immunodeficiency virus and amyloidosis do not have long-term survival (Levy et al., 2006).
Patients with idiopathic DCM have variable natural history. Some patients may be stable
over years or even decades. This stability may be in response to pharmacological or device
220 Advances in the Diagnosis of Coronary Atherosclerosis

therapy or it can be due to spontaneous reverse remodeling. On the other hand, the clinical
stability of some patients deteriorates suddenly and the hemodynamic situation requires
inotropic or mechanical support (Teuteberg et al.,2006).

2.2 Etiology
DCM is the major cause of congestive heart failure (CHF).Other most frequent specific
Cardiomyopathies causing CHF include ischemic or hypertensive cardiomyopathies and
nonsystolic heart failure (Owan et al.,2006).In DCM after excluding the specific disease
about 50 percent is called idiopathic DCM( Felker et al., 2000).However, some of these
idiopathic DCM cases are caused by genetic abnormalities or some environmental etiologies
that are not diagnosed at the time of primary presentation and with more imaging or
molecular evaluation, many of these idiopathic cases will have a specific etiology.

2.3 Specific forms of dilated cardiomyopathy


There are many important causes of secondary CMP including alcohol, HIV infection,
metabolic impairments and cardiotoxic drugs such as doxorubicin. There are four specific
causes of dilated CMP with more probability of recovery.

2.3.1 Tachycardia-induced cardiomyopathy


Patients with recurrent or permanent tachycardia such as atrial fibrillation or
supraventricular tachycardia may develop CHF or DCM. Atrial fibrillation is a frequent
disease among patients affected by CHF (Ghaemian et al., 2011).There is a high probability
of recovery by controlling the arrhythmia (Redfield et al., 2000).Thus , patients should be
treated to restore normal sinus rhythm.

2.3.2 Peripartum cardiomyopathy


This form of cardiomyopathy presents between 6 months to the last month of pregnancy
(Elkayam et al., 2001).Usually the prognosis is good. However, women who recover the first
attack are at increased risk of recurrence during the next pregnancies.

2.3.3 Broken heart or tako-tsubo syndrome


This CMP is usually caused by a stressful condition. This CMP is more common in women
and the patients usually recover with treatment. Electrocardiogram may show findings of
myocardial infarction, but in coronary angiography epicardial coronary arteries do not show
stenosis.

2.3.4 Alcoholic cardiomyopathy


This form of CMP usually is related to the amount of consumption of alcohol with recovery
by cessation of alcohol. The mechanism usually is direct toxicity of the alcohol (Fernandez-
sola et al.,2006).This CMP is more common in men, patients usually have a history of
excessive consumption of alcohol for more than 10 years. Patients may have a fulminant
course with biventricular failure. Alcohol may precipitate some arrhythmias including atrial
fibrillation or supraventricular tachycardia. Many electrocardiographic abnormalities
including QT interval prolongation are common. Hemodynamic and echocardiographic
findings are similar to those of DCM patients. Cessation of consumption of the alcohol is the
main component of treatment.
Cardiac Catheterization and Coronary Angiography in Patients with Cardiomyopathy 221

2.4 Clinical presentation of the dilated cardiomyopathy


DCM is more frequent in middle age man, but it affects all ages. The incidence if ischemic
CMP is higher than DCM. The clinical presentation of patients with DCM is variable. While
some patients gradually become symptomatic, others may develop symptoms over only
weeks and yet some of them have fulminant course of congestive heart failure (McCarthy et
al., 2000).The most frequent symptoms of patients are dyspnea and fatigue.

2.5 Laboratory evaluation


Screening the biochemical testing including serum electrolytes, calcium, phosphorus and
serum creatinine is necessary. Rheumatological tests such as antinuclear antibody (ANA)
and erythrocyte sedimentation rate should also be obtained. Endocrine tests including
thyroid function test and urinary examination of catecholamine levels may be helpful.
Serum troponin levels to assess myocardial necrosis and brain natriuretic peptide levels are
usually obtained.

2.5.1 Electrocardiography
Nonspecific electrocardiographic changes include sinus tachycardia, interventricular
conduction delay and poor R progression and left bundle branch block. Other
electrocardiographic abnormalities such as ST segment and T wave abnormalities may also
be seen. Patients may show a wide spectrum of supraventricular and ventricular
arrhythmia.

2.5.2 Echocardiography
By two-dimensional echocardiography ventricular size and function are usually evaluated.
Doppler echocardiography is helpful to evaluate valvular stenosis or regurgitation and
restrictive filling patterns. Dobutamine stress echocardiography is helpful to differentiate
ischemic from idiopathic DCM and also to assess contractile reserve of myocardium.

2.5.3 Radionuclide imaging


This imaging technique is useful to identify ischemic CMP and also can assess cardiac
function and ejection fraction of both ventricles.

2.5.4 Magnetic Resonance Imaging (MRI)


Cardiac MRI is useful to diagnose some specific CMPs such as myocarditis, amyloidosis,
sarcoidosis and infiltrative and inflammatory cardiomyopathies and arrhythmogenic right
ventricular dysplasia (Maceira et al., 2005; Mahrholdt et al.,2004; Tandri et al., 2005).

3. Ischemic cardiomyopathy
When myocardial dysfunction is secondary to occlusion of the coronary arteries, the term
ischemic CMP is used and is the most common cause of heart failure. Based on the
pathophysiological processes ischemic CMP may be classified into three groups. The first
one is called myocardial hibernation. In this form of ischemic CMP a reduced coronary flow
will result in persistent contractile dysfunction and revascularization procedures will
usually restore myocardial function, at least partially. Myocardial stunning is the second
form of ischemic CMP. In this form the contractile dysfunction of the viable myocardium is
transient and is usually caused by reduced sensitivity of contractile proteins to calcium.
222 Advances in the Diagnosis of Coronary Atherosclerosis

Finally in the third type, the contractile dysfunction is secondary to irreversible cell death of
the myocardium.

3.1 Revascularization in ischemic CMP


In patients with ischemic CMP revascularization is beneficial only when the myocardium is
viable and revascularization procedures cause an improvement in left ventricular function.
In patients with severe left ventricular dysfunction, but with viable myocardium, coronary
artery revascularization results in an average 8 to 10 percent of improvement in left
ventricular ejection fraction (Kumpati et al.,2001).This will be associated with improvement
in clinical situation and survival of the patients. However, if the viable myocardium is not
considerable improvement of left ventricular function will not occur. It has been reported
that 20 to 25 percent or more of the myocardium is the threshold of myocardial viability for
which surgery is indicated (De Bonis & Alfieri 2006).Patients with ischemic CMP should be
examined for detection of myocardial viability.Thallium-201 single-photon emission
computed tomography (SPECT) and dobutamine stress echocardiography are usually used
for this purpose. When both of these two methods predicated viability, the global left
ventricular ejection fraction rose from 35 to 40 percent after operation (Starling et al.,
2003).In other reports positron emission tomography (PET) was better than SPECT to
predict improvement of left ventricular ejection fraction after revascularization (Pagano et
al., 1999;Starling et al., 2003). In a meta-analysis of 24 studies in patients with ischemic CMP
revascularization decreased the risk of death by 79.6 percent in patients with viable
myocardium detected by SPECT, dobutamine echocardiography or positron emission
tomography (Bishay et al., 2000). Thus, in patients with heart failure secondary to coronary
artery disease coronary angiography and viability study should be performed.

4. Right and left heart catheterization


Catheterization of the right and left side of the heart are usually performed to examine the
hemodynamics of the heart i.e. to measure intracardiac pressure, cardiac outputs, and vascular
resistances. In patients with heart failure, respiratory failure, valvular heart disease, pericardial
disease, restrictive or hypertrophic cardiomyopathies hemodynamic assessment is of great
value for the diagnosis and management of the patients. If necessary angiographic procedures
such as left or right ventriculography, pulmonary angiography, and aortography may be
performed during cardiac catheterization. Besides, selective angiographic procedures include
native coronary, coronary bypass graft and carotid angiography.

4.1 Hemodynamic evaluation


Catheterization laboratories use disposable, fluid – filled transducers to measure pressure.
For hemodynamic evaluation, the right heart catheter is placed in the pulmonary artery and
a pigtail catheter in the aorta. Cardiac out is usually measured by the thermodilution
method. Oxymetric analysis is performed on the blood collected from the aorta and the
pulmonary artery, cardiac output is calculated by the Fick method , the presence of a left- to
- right shunt is also screened. Catheter of the aorta is advanced into the left ventricle and the
right- sided catheter into pulmonary capillary wedge position and the pressures are
measured simultaneously. Then the right sided catheter is withdrawn and all the pressures
are recorded. With the simultaneous measurement of the pressure of right and left ventricles
it is possible to screen for restrictive or constrictive physiology. Then by pulling back the
left- sided catheter the pressure gradient across the aortic valve is determined.
Cardiac Catheterization and Coronary Angiography in Patients with Cardiomyopathy 223

4.2 Complications
Cardiac catheterization also is a safe procedure, potentially has some complications. In
patients with left ventricular thrombus or aortic valve endocarditis it is better to avoid
inserting catheters into the left heart. Catheter induced arrhythmia is another potential
complication of right or left heart. Right or left bundle branch blocks may occur if the
catheter is placed in the right ventricular outflow tract or crossed the aortic valve,
respectively. Thus, patients with right or left bundle branch block are at potential risk of
developing complete heart block during cardiac catheterization. Renal failure, contrast
reactions, vascular complications are the more common complications of cardiac
catheterization. The incidence of cerebrovascular accident, myocardial infarction, and death
is less than 1 percent. Other rare complications include infection, chamber or vessel
perforation, cholesterol embolism, air embolism, and radiation -induced skin injury.

4.3 Right heart catheterization


Right heart catheterization is performed from the right internal Jugular, femoral or subclavian
veins using a balloon flotation catheter. For right heart catheterization fluoroscopy is not
necessary and by attaching the catheter to a pressure transducer the operator can advance the
catheter. Pressure is recorded in the right sided chambers and the pulmonary artery. With the
catheter in the pulmonary artery, it is possible to measure thermodilution cardiac output and
perform oximetry to detect a shunt and determine cardiac output by the Fick method. The
catheter also can be advanced to a pulmonary capillary wedge position to obtain the wedge
pressure. If by the pressure-guided technique catheter does not advance successfully and also
when the right heart catheterization is performed from the femoral veins fluoroscopy is used.
The normal pressure of the right atrium is 2 to 6 mmHg with positive “a” and “v” waves and
negative “x” and “y” descents (Fig.1).

Fig. 1. Normal right atrial waveform


The atrial contraction increases the right atrial pressure producing the “a” wave. At the
initial part of ventricular systole the downward motion of the tricuspid valve and the atrial
relaxation produce the “x” descent. An inconsistent “c” waves after the “a” represents the
224 Advances in the Diagnosis of Coronary Atherosclerosis

motion of the tricuspid annulus toward the right atrium at the onset of the ventricular
systole. The distance of the “c” from the “a” is the same as the PR interval in the
electrocardiogram and first degree atrioventricular block produces a more visible “c” wave.
The “x” descent represents the decrease of the right atrial pressure in ventricular systole
which is followed by the “v” wave in atrial diastole when the right atrium is filled by
venous return. The peak of the “v” wave occurs at the end of ventricular systole when the
right atrium is maximally filled and is at the same time of T wave on the electrocardiogram.
The final “y” descent represents the rapid emptying of the right atrium at the initial part of
ventricular diastole when the tricuspid valve opens. As shown in Figure 2 the normal right
ventricular systolic pressure is 20 to 30mmHg, and normal right ventricular end-diastolic
pressure is 0 to 8 mmHg. The right ventricular pressure normally consists of a rapid rise of
pressure followed by a decline during ventricular contraction and then rapid downward
motion with ventricular relaxation. At the beginning of diastole the pressure is low and with
ventricular filling increases gradually.

Fig. 2. Normal right ventricular waveform


The normal range of pulmonary artery systolic pressure is about 20 to 30 mmHg while the
diastolic pressure is 4 -12 mmHg . As is shown in Figure 3 the pressure waveform of the
pulmonary artery increase rapidly during systole and reaches a peak. However, during
diastolic descent of the pressure there is a dicrotic notch.

Fig. 3. Normal pulmonary artery waveform


Cardiac Catheterization and Coronary Angiography in Patients with Cardiomyopathy 225

During right heart catheterization the pulmonary capillary wedge pressure (PCWP)
waveform is usually evaluated. The pressure waveform is better to obtain with an end-
hole catheter. The end-hole catheter obstructs the antegrade flow into the pulmonary
artery, so it reflects the left atrial pressure, or the PCWP. This pressure can be obtained by
a Swan-Ganz catheter in the pulmonary artery. By inflating the balloon of this catheter
and obstructing the flow, the PCWP is obtained. The normal mean pressure of the PCWP
is about 2 to 14mmHg and consists of positive “a” and “v” and negative “x” and “y”
waves (Fig.4).

Fig. 4. Normal pulmonary capillary wedge pressure waveform


However, “c” wave which is sometimes seen in an atrial pressure is usually not seen in
PCWP and also because the PCWP is the pressure transmitted through the pulmonary
capillary bed, a time delay occurs between an electrocardiographic event and the onset of
the relevant wave. For example the “a” wave of the right atrial pressure follows the P wave
of the electrocardiogram by about 80 milliseconds while it is about 240 milliseconds in
PCWP. Also, the peak of the “v” wave in PCWP tracing occurs after the T wave. Finally it
should be noted that the oxygen saturation obtained from the catheter in the PCWP position
is greater than 90%.

4.4 Left heart catheterization


Left heart catheterization is usually performed using a pigtail catheter. The catheter is
advanced to the ascending aorta and then is positioned in the left ventricle. For this purpose,
it may be necessary to rotate the catheter clockwise or counterclockwise gently to cross the
valve. Occasionally a J- tipped stiff guide wire should be used.
The left ventricular waveform has a rapid upstroke during ventricular contraction and then
during relaxation the pressure decreases (Fig.5).
At the initial part of the diastole the ventricular pressure is low and increases slowly until
the end diastolic portion and the normal diastolic pressure is 8 to 16 mmHg. Left ventricular
end -diastolic pressure is the pressure just before the abrupt increase in systolic pressure.
Central aortic pressure waveforms normally have a rapid upstroke, a systolic peak followed
by a dicrotic notch due to closure of the aortic valve (Fig.6).
226 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 5. Prominent “a” wave in left ventricular waveform

Fig. 6. Normal central aortic waveform


During hemodynamic evaluation for screening for the presence of a restrictive or
constrictive physiology, simultaneous measurement of left and right ventricular pressures is
performed. Normally peak systolic pressure changes during expiration and inspiration in
the right and left ventricle parallel each other (Fig.7) and the diastolic pressures of the two
ventricles differ by at least 5 mmHg during the respiratory cycle (Fig.8).

4.5 Hemodynamic calculations


During cardiac catheterization and hemodynamic evaluation, hemodynamic calculations are
made. For example, calculation of the cardiac output by the Fick method, calculations of the
pulmonary and peripheral vascular resistances, and calculations of the mitral and aortic
valves areas can be obtained. However, most of the computerized hemodynamic data
collection systems have the softwares to calculate these values automatically.

4.6 Trans - septal heart catheterization


In some situations it is not possible to enter the left ventricle via retrograde approach. For
example, in the presence of a mechanical aortic valve at severe aortic stenosis it may be
necessary to measure left ventricular pressure or perform left ventriculography and trans -
Cardiac Catheterization and Coronary Angiography in Patients with Cardiomyopathy 227

septal approach can be helpful. During some interventional procedures such as mitral
balloon valvuloplasty, percutaneous mitral valve repairs with clips, some electrophysiologic
procedures, and placement of a percutaneous left ventricular assist device may require trans
-septal puncture.

Fig. 7. Normal concomitant waveform of right ventricle and left ventricular in systole

Fig. 8. Normal concomitant right and left ventricular waveform in diastole


During a trans-septal puncture the goal is usually to puncture the atrial septum at the fossa
ovalis and the procedure is performed from the right femoral vein. At the same time a
pigtail catheter is positioned on the aortic valve to identify the aorta by fluoroscopy. The
puncture of the interatrial septum is performed using the Brockenbrough needle inserted
into a dilator. After the puncture, a guidewire is advanced into the left atrium and the
dilator is advanced over the wire in order to position the sheath within the left atrium and
left atrial pressure is recorded. Then based on the procedure, the rest of the catheterization
or intervention can be performed.
228 Advances in the Diagnosis of Coronary Atherosclerosis

5. Abnormal hemodynamic waveforms in patients


In patients with left ventricular systolic dysfunction including dilated CMP the
hemodynamic findings are nonspecific. Heart failure increases the pulmonary capillary
wedge pressure and right-sided pressures. Chronic heart failure increases the pulmonary
artery pressure because of reactive and fixed increases in pulmonary vascular resistance,
leading to pulmonary hypertension and secondary tricuspid regurgitation and right heart
failure. In patients with systolic dysfunction left ventricular end-diastolic pressure is
increased and a prominent “a” wave may be seen (Fig 9).

Fig. 9. Pressure waveform in a patient with left ventricular dysfunction showing a


prominent “a” wave
In patients with advanced stages of left ventricular dysfunction, left ventricular pressure
may have a delay to reach the systolic peak due to diminished contractility (Fig 10).

Fig. 10. Left ventricular pressure waveform in a patient with severe left ventricular
dysfunction which have a triangular appearance
Another presentation of severely reduced left ventricular contractility is the appearance of
pulsus alternans on the aortic or left ventricular pressure waveform (Fig 11).
Ambulatory patients with dilated CMP may have a relatively low normal resting cardiac
output and a modest elevation in both right-and left-sided filling pressures. However, in
advanced stages of heart failure, the systemic vascular resistance rises significantly in response
to the reduced cardiac output. Also, in advanced heart failure in dilated CMP patients
Cardiac Catheterization and Coronary Angiography in Patients with Cardiomyopathy 229

biventricular failure is usually characteristic. The right atrial pressure waveform will
demonstrate steep “x” and “y” descents indicative of severe volume overload and right
ventricular systolic and diastolic dysfunction. Kussmaul sign, i.e., absence of the normal
respiratory fall in the right atrial pressure is common and as a result of tricuspid regurgitation
the “y” descent is steep. The PCWP waveform may demonstrate a prominent “v” wave. The
left ventricular pressure waveform is characterized by an elevation in pressure throughout
early diastole. The arterial tracing may show a narrow pulse pressure (pulsus parvus).

Fig. 11. Aortic pressure tracing in a patient with severe left ventricular dysfunction pulsus
alternans
In patients with right-sided heart failure similar to left heart failure, pulsus alternans may be
present in right-sided pressure waveforms (Fig 12).

Fig. 12. Pulsus alternans in the right ventricular wave form in a patient with right-sided
heart failure
In the case of tricuspid regurgitation secondary to pulmonary hypertension as in primary
tricuspid regurgitation a prominent “v” wave on the right atrial waveform may appear (Fig
13) and in severe cases of tricuspid regurgitation ventricularization of the right atrial
waveform may occur (Fig 14).
230 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 13. Prominent “v” wave in the right atrial pressure in a patient with severe tricuspid
regurgitation

Fig. 14. Ventricularization of the right atrial waveform in a patient with severe tricuspid
regurgitation
In patients with hypertrophic obstructive cardiomyopathy the systolic anterior motion of
the mitral valve obstructs the left ventricular outflow tract causing a pressure gradient
between the left ventricle and aorta (Fig 15). And the aorta pressure waveform may show
the characteristic pattern of spike and dome configuration (Fig 16). In some cases the
gradient may be absent at rest but can be provoked by decreasing the preload.

Fig. 15. Simultaneous left ventricular and aortic pressure waveform in a patient with
hypertrophic obstructive cardiomyopathy which shows significant pressure gradients at rest
Cardiac Catheterization and Coronary Angiography in Patients with Cardiomyopathy 231

Fig. 16. Aortic pressure waveform showing “spike-and-dome” configuration

6. Left ventriculography
Left ventriculography is performed during a left heart catheterization to assess left
ventricular function. Left ventriculography can also be helpful to identify the presence of
mitral regurgitation, ventricular septal defect, and left ventricular hypertrophy. In patients
with high pulmonary capillary wedge pressure or left ventricular end-diastolic pressure the
injection of contrast may impose the patient to the risk of acute pulmonary edema. In
addition, in patients with a mobile left ventricular thrombus seen on echocardiography it is
better not to perform left ventriculography. Left ventriculography is commonly performed
by a power injector and a pigtail catheter positioned in the mid ventricle.
For assessment of ventricular systolic function, ejection fraction is calculated via left
ventriculography. Usually computer-based software is applied and the left ventricular end-
diastolic and end-systolic contours are traced. Diastolic and systolic areas can be determined
and based on the assumptions regarding the geometry of the heart, ventricular volumes are
calculated and then the left ventricular ejection fraction can be calculated. Furthermore, in
patients with coronary artery disease, regional left ventricular function can be assessed.
Based on the distribution of the coronary arteries the left ventricle is divided into some
segments. The contractions of the segments are evaluated. If the contraction of each segment
is reduced it is classified as “hypokinetic” and if there is no contraction it is called
“akinetic”. Finally, if during systole the segment moves outward it is called “dyskinetic”. In
patients with hypertrophic cardiomyopathy, because of excessive hypertrophy, in left
ventriculography cavity obliteration and a small ventricular end-systolic volume are
commonly seen. In these patients if ventricular outflow tract obstruction also is present,
systolic anterior motion of the mitral valve can be noted. Interestingly, in apical form of
hypertrophic cardiomyopathy the ventriculogram appears with a “spade” – shaped contour.
In patients with apical ballooning syndrome who have transient severe left ventricular
dysfunction in the absence of coronary artery disease left ventriculography shows aneurysm
of the left ventricle which predominantly affects the apex of the heart.

7. Right ventriculography
Right ventriculography is usually indicated in congenital heart disease. However, right
ventriculography in adults may be helpful in the diagnosis of right ventricular dysplasia
and determination of right ventricular systolic function. In patients with arrhythmogenic
right ventricular dysplasia several findings associated with this disorder have been noted.
However, there is no pathognomonic feature in right ventriculography in these patients.
232 Advances in the Diagnosis of Coronary Atherosclerosis

8. Coronary angiography
Coronary angiography is considered the gold standard for diagnosing coronary artery
disease and can be useful to differentiate ischemic from dilated forms of cardiomyopathies.
During coronary angiography the dominant vessel and also the vascular territories for each
artery can be identified. Obstructive coronary artery disease is usually defined by
angiography when a greater than 50% diameter narrowing is seen. When the lesions narrow
the lumen by more than 70%, they are generally believed to be hemodynamically significant.
However, there are some limitations regarding the usefulness of coronary angiography in
diagnosing coronary artery disease. Coronary angiography only assesses the arterial lumen,
while atherosclerosis is a disease of the arterial wall. There may be a substantial amount of
plaque in the arterial wall before it is evident by angiography. Thus, a normal coronary
arteriogram does not exclude the presence of coronary artery disease. Second, there is a
wide range of interobserver variability in interpreting the coronary arteriogram of a patient.
Third, depending on the image quality, the operator technique, patient body habitus, the
vessel tortuosity, and the presence of overlapping segments, the ability of the angiography
to assess a lesion may be affected. Finally, for identification of a luminal narrowing a
reference normal segment of the vessel is usually used for comparison. In patients with
diffuse coronary lesions, a true normal segment is not present and the arterial lumen may
appear uniform.
Ischemic cardiomyopathy results following severe coronary artery disorders and usually
angiography must be performed for proper diagnosis (Shokrzadeh et al., 2008). Coronary
atherosclerosis usually causes narrowing of the lumen of the coronary arteries in advanced
stages. In the early stages of atherosclerosis, compensatory enlargement of the artery
maintains the coronary artery lumen. Thus, coronary angiography which only assesses the
arterial lumen can not detect the early stages of atherosclerosis. However, techniques such
as intravascular ultrasound provide images of arterial wall and detect coronary
atherosclerosis in earlier stages. Coronary atherosclerosis can be detected anywhere along
the course of the artery.
There are many indications for performing coronary angiography. For example, in different
clinical situations such as myocardial infarction, acute coronary syndromes stable angina,
patients with valvular or congenital heart diseases coronary angiography may be useful to
define coronary artery anatomy. However, coronary angiography should not be performed
in patients who do not want to undergo revascularization, patients with severe left
ventricular dysfunction in whom revascularization is not likely to improve ventricular
function, patients with acute bleeding, unexplained fever, and in patients with severe
coagulopathy.

9. Conclusion
Cardiac catheterization is performed in patients with heart failure to assess the etiology, to
evaluate the hemodynamic status of the patient, and to define therapeutic responses. While
the major part of the hemodynamic evaluation is assessed in the supine state, exercise
conditions can also be evaluated. After hemodynamic evaluation of the patient angiography
should be performed to define the coronary anatomy and if needed left ventriculography to
assess ventricular contractility. Ventriculography is also helpful to define ventricular size
and mitral regurgitation, although most patients will have had complete echocardiographic
study prior to catheterization. In the absence of coronary artery disease to explain
Cardiac Catheterization and Coronary Angiography in Patients with Cardiomyopathy 233

ventricular dysfunction, an endomyocardial biopsy should be considered for a specific


diagnosis.

10. Acknowledgement
I wish to thank Dr Safoora Masoumi for her help in providing this chapter.

11. References
Ammash, NM., Seward, JB., Bailey, KR., Edwards, WD., & Tajik, AJ. (2000).Clinical profile
and outcome of idiopathic restrictive cardiomyopathy. Circulation, Vol. 101, No.21,
(may 2000), pp.2490-2496, ISSN 0009-7322
Bishay, ES., McCarthy, PM., Cosgrove, DM., Hoercher, KJ., Smedira, NG., Mukherjee, D.,
White, J., & Blackstone, EH.(2000). Mitral valve surgery in patients with severe left
ventricular dysfunction. Eur J Cardiothorac Surg, Vol.17, No.3, (Mar 2000), pp.213-
221, ISSN 1010-7940
De Bonis, M., & Alfieri, O.(2006). Surgery insight: Surgical methods to reverse left
ventricular remodeling. Nat Clin Pract Cardiovasc Med, Vol.3, No.9, (Sep 2006),
pp.507-513, ISSN 1759-5002
Dec, GW., & Fuster, V.(1994). Idiopathic dilated cardiomyopathy. N Engl J Med, Vol. 331,
No.23, (Dec 1994), pp.1564-1575, ISSN 0028-4793
Elkayam, U., Tummala, PP., Rao, K., Akhter, MW., Karaalp, IS., Wani, OR., Hameed, A.,
Gviazda, I., & Shotan A.(2001) Maternal and fetal outcomes of subsequent
pregnancies in women with peripartum cardiomyopathy. N Engl J Med, Vol.344,
No.21(May 2001), pp.1567-1571, ISSN 0028-4793
Felker, GM., Thompson, RE., Hare, JM., Hruban, RH., Clemetson, DE., Howard, DL.,
Baughman, KL., & Kasper, EK. (2000) Underlying causes and long-term survival in
patients with initially unexplained cardiomyopathy. N Engl J Med, Vol.342, No.15,
(Apr 2000), pp.1077-1084, ISSN 0028-4793
Fernández-Solà, J., Fatjó, F., Sacanella, E., Estruch, R., Bosch, X., Urbano-Márquez, A., &
Nicolás, JM.(2006) Evidence of apoptosis in alcoholic cardiomyopathy. Hum Pathol,
Vol.37, No.8(Aug 2006), pp.1100-1110, ISSN 0046-8177
Ghaemian, A., Salehifar, E., Jalalian, R., Ghasemi, F., Azizi, S., Masoumi, S., Shiraj, H.,
Mohammadpour, RA., & Bagheri GA.(2011). Zinc and Copper Levels in Severe
Heart Failure and the Effects of Atrial Fibrillation on the Zinc and Copper Status.
Biol Trace Elem Res, [ Epub ahead of print], (2011 Jan), DOI 10.1007/s12011-011-
8956-6
Kumpati, GS., McCarthy, PM., & Hoercher, KJ.(2001). Surgical treatments for heart failure.
Cardiol Clin, Vol.19, No .4, (Nov 2001), pp. 669-681, ISSN 0160-9289
Levy, WC., Mozaffarian, D., Linker, DT., Sutradhar, SC., Anker, SD., Cropp, AB., Anand, I.,
Maggioni, A., Burton, P., Sullivan, MD., Pitt, B., Poole-Wilson, PA., Mann, DL., &
Packer, M.(2006) The Seattle heart failure model-Prediction of survival in heart
failure.Circulation, Vol.113, No.11, ( Mar 2006), pp.1424-1433, ISSN 0009-7322
Maceira, AM., Joshi, J., Prasad, SK., Moon, JC., Perugini, E., Harding, I., Sheppard, MN.,
Poole-Wilson, PA., Hawkins, PN., & Pennell DJ. (2005).Cardiovascular magnetic
resonance in cardiac amyloidosis. Circulation, Vol .111, No.2, (Jan 2005), pp.186-193,
ISSN 0009-7322
Mahrholdt, H., Goedecke, C., Wagner, A., Meinhardt, G., Athanasiadis, A., Vogelsberg, H.,
Fritz, P., Klingel, K., Kandolf, R., & Sechtem U.(2004) Cardiovascular magnetic
234 Advances in the Diagnosis of Coronary Atherosclerosis

resonance assessment of human myocarditis-A comparison to histology and


molecular pathology. Circulation, Vol. 109, No.10, (Mar 2004), pp. 1250-1258, ISSN
0009-7322
Maron, BJ., Towbin, JA., Thiene, G., Antzelevitch, C., Corrado, D., Arnett, D., Moss, AJ.,
Seidman, CE., Young, JB., American Heart Association., Council on Clinical
Cardiology., Heart Failure and Transplantation Committee., Quality of Care and
Outcomes Research and Functional Genomics and Translational Biology
Interdisciplinary Working Groups., & Council on Epidemiology and
Prevention.(2006). Contemporary definitions and classification of the
cardiomyopathies—An American Heart Association Scientific Statement from the
Council on Clinical Cardiology, Heart Failure and Transplantation Committee;
Quality of Care and Outcomes Research and Functional Genomics and
Translational Biology Interdisciplinary Working Groups; and Council on
Epidemiology and Prevention. Circulation, Vol.113, No.14, (Apr 2006), pp 1807-
1816, ISSN 0009-7322
McCarthy, RE., Boehmer, JP., Hruban, RH., Hutchins, GM., Kasper, EK., Hare, JM., &
Baughman KL.(2000). Long-term outcome of fulminant myocarditis as compared
with acute (nonfulminant) myocarditis. N Engl J Med, Vol.342, No.10, (Mar 2000),
pp. 690-695, ISSN 0028-4793
Owan, TE., Hodge, DO., Herges, RM., Jacobsen, SJ., Roger, VL., & Redfield MM.(2006)
Trends in prevalence and outcome of heart failure with preserved ejection fraction.
N Engl J Med, Vol.355, No.3, (Jul 2006), pp.251-259, ISSN 0028-4793
Pagano, D., Bonser, RS., & Camici, PG. (1999) .Myocardial revascularization for the
treatment of post-ischemic heart failure. Curr Opin Cardiol, Vol.14, No.6, (Nov
1999), pp.506-509, ISSN 0268-4705
Redfield, MM., Kay, GN., Jenkins, LS., Mianulli, M., Jensen, DN., & Ellenbogen, KA.(2000)
Tachycardia-related cardiomyopathy: A common cause of ventricular dysfunction
in patients with atrial fibrillation referred for atrioventricular ablation. Mayo Clinic
Proc, Vol.75, No.8(Aug 2000), pp.790-795, ISSN 0025-6196
Salehifar, E., Shokrzadeh, M., Ghaemian, A., Aliakbari, S., & Saeedi Saravi, SS.(2008). The
study of Cu and Zn serum levels in idiopathic dilated cardiomyopathy (IDCMP)
patients and its comparison with healthy volunteers. Biol Trace Elem Res, Vol.125,
No.2, ( Nov 2008), pp.97-108 ISSN 0163-498
Shokrzadeh, M., Ghaemian, A., Salehifar, E., Aliakbari, S., Saravi, SS., & Ebrahimi, P.(2009).
Serum zinc and copper levels in ischemic cardiomyopathy. Biol Trace Elem Res,
Vol.127, No.2, (Feb 2009), pp.116-123, ISSN 0163-498
Starling, RC., McCarthy, PM., & Yamini, MH.(2003). Surgical treatment of chronic
congestive heart failure, In: Heart Failure: A Companion to Braunwald's Heart Disease,
D.L. Mann, (Ed.), pp.717-736, WB Saunders, ISBN 0721694454 Philadelphia
Tandri, H., Saranathan, M., Rodriguez, ER., Martinez, C., Bomma, C., Nasir, K., Rosen, B.,
Lima, JA., Calkins, H., & Bluemke DA.(2005). Noninvasive detection of myocardial
fibrosis in arrhythmogenic right ventricular cardiomyopathy using delayed-
enhancement magnetic resonance imaging. J Am Coll Cardiol, Vol.45, No.1, (Jan
2005), pp.98-103, ISSN 0735-1097
Teuteberg, JJ., Lewis, EF., Nohria, A., Tsang, SW., Fang, JC., Givertz, MM., Jarcho, JA.,
Mudge, GH., Baughman, KL., & Stevenson LW.(2006) Characteristics of patients
who die with heart failure and a low ejection fraction in the new millennium. J
Cardiac Fail, Vol .12, No. 1, (Feb 2006), pp.47-53, ISSN 071-9164
13

Contrast-Induced Nephropathy in Patients


with Type 2 Diabetes Mellitus and
Coronary Artery Disease: Update
and Practical Clinical Applications
Richard E. Katholi and Charles R. Katholi
Southern Illinois University School of Medicine and
University of Alabama at Birmingham School of Medicine
USA

1. Introduction
Contrast-induced nephropathy (CIN) is an injury to the kidney as a result of exposure to
intravascular iodinated contrast medium. It represents an increasing health care burden and
challenge as the frequency of diagnostic imaging and interventional studies increase,
particularly among populations at risk of developing CIN. As the population ages,
decreased renal function and increased atherosclerotic cardiovascular disease become more
prevalent. An increasing incidence of obesity with resultant metabolic syndrome and/or
type 2 diabetes mellitus also increases the population at risk for CIN (Toprak et al., 2006).
Coronary artery disease is a major complication of type 2 diabetes mellitus. Thus, patients
with type 2 diabetes mellitus often require coronary angiography and coronary intervention
and are at risk of CIN.

2. Contrast-induced nephropathy in patients with type 2 diabetes mellitus


and coronary artery disease
For clinical and research purposes, CIN is defined as an acute decline in renal function (rise
in serum creatinine by 25% or greater than 0.5 mg/dL from baseline or fall in estimated
glomerular filtration rate (eGFR) by greater than 25%) after systemic contrast medium
administration in the absence of other causes. Typically, CIN onset occurs within 24-48
hours of exposure, serum creatinine levels peak in 3-5 days, and renal function returns to
baseline in 7-21 days. If renal function does not return to baseline, other causes of acute
renal injury, like atheroembolism or hypotension, should be suspected (Saleem et al., 1996).
CIN is associated with both short-and long-term adverse outcomes, including the need for
renal replacement therapy, increased length of hospital stay, major cardiac adverse events,
and mortality. The incidence of CIN is less than 5% in patients with normal renal function
and 15-50% in patients with baseline renal dysfunction (eGFR less than 60 mL/min/1.73m2).
The increased risk of CIN in diabetic patients with reduced renal function is well
documented (Parfrey et al., 1989). Yet, diabetic patients without overt renal dysfunction are
also at risk as compromised renal function may not manifest until an acute renal insult
236 Advances in the Diagnosis of Coronary Atherosclerosis

results from the administration of contrast medium. CIN carries short-term and long-term
mortality. Acute renal failure after coronary intervention is associated with a 36% in-
hospital mortality rate and a 19% 2-year survival rate. Thus, understanding why patients
with type 2 diabetes are at increased risk of CIN and controlling factors to decrease the risk
of CIN, has major short- and long-term benefits to patients with type 2 diabetes (Nikolsky et
al., 2004). Most clinical studies on CIN have mainly been in patients with type 2 diabetes
mellitus rather than type 1 diabetes mellitus and, thus, the remarks in this chapter will be
confined to patients with type 2 diabetes kidney disease.

2.1 Mechanisms of contrast-induced nephropathy in patients with type 2 diabetes


mellitus
The pathogenesis of CIN is complex with a cascade of contributing factors that are not fully
understood. Factors contributing to CIN in patients with type 2 diabetes mellitus include
decreased GFR, endothelial dysfunction and increased reactivity to intrarenal adenosine
(Pfueger et al., 2000). After injection of contrast medium, renal blood flow increases
transiently, followed by a more prolonged decrease, particularly at the corticomedullary
junction of the kidney. The outer medulla is particularly susceptible to ischemic injury
because of its high metabolic activity and low prevailing oxygen tension. Associated with
the decrease in renal blood flow, there is a decrease in glomerular filtration rate due to
afferent renal arteriolar vasoconstriction which is calcium-dependent with increased
intrarenal adenosine and increased endothelin-1 activity as likely mediators of the
vasoconstriction (Arakawa et al., 1996). The risk of CIN increases if there are inadequate
compensatory vasodilatory responses, such as prostaglandins (E2 and I2) and nitric oxide.
Renal tubular cellular injury, in part, is mediated by generation of oxygen-free radicals.
Intrarenal adenosine accumulates due to the depletion of adenosine triphosphate as a
consequence of proximal tubular stress due to osmotic load and the large size of contrast
media molecules. The renal toxicity from the direct effects of contrast media is reversible. It
has been shown in vitro that renal tubular cells respond to contrast media exposure by
increasing the concentrations of extracellular adenosine and by decreasing the activity of
mitochondrial enzymes without altering viability. A late effect of intrarenal adenosine is
oxygen-free radical production due to the catabolism of intrarenal adenosine to xanthine.
A role for intrarenal adenosine as a renal vasoconstrictor and substrate for oxygen-free
radical formation is supported in human studies. Adenosine increases in urine following
contrast medium; the magnitude of adenosine release and depression of creatinine clearance
is proportional to the osmolality of the contrast agent, essentially a dose response
relationship. Further, an inhibitor of adenosine uptake, dipyridamole, exacerbates the fall in
GFR after contrast medium. In addition, blockade of the renal arterial adenosine receptors
with theophylline attenuates the fall in GFR following contrast medium. Finally, following
pretreatment with allopurinol (a xanthine oxidase inhibitor), urinary xanthine increases and
the fall in GFR after contrast medium is lessened.
Endothelin is a potent renal afferent arteriolar vasoconstrictor, like intrarenal adenosine.
Intravascular administration of contrast medium induces exacerbated release of urinary
endothelin in patients with impaired renal function. Human studies show no significant
increase in plasma endothelin levels until a volume of contrast media administered is
greater than 150 mL (Clark et al., 1997). However, in diabetic patients or in patients with
renal insufficiency, significant endothelin levels are detected when less than 100 mL of
contrast medium are administered intraarterially.
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 237

In patients with normal GFR, the risk of CIN is likely less, not only because of more rapid
clearance of contrast medium from the kidney (less time for generation of oxygen-free
radicals), but, presumably, because of the presence of a variety of endogenous vasodilators
that protect against renal ischemia, including prostaglandins (E2 and I2), atrial natriuretic
peptide and nitric oxide. In patients with GFR of less than 60 mL/min/1.73 m2, the risk of
CIN increases with prolonged clearance of contrast medium.
In summary, patients with type 2 diabetes mellitus appear to be at increased risk of CIN, not
only because chronic renal disease is common in these patients, but also because there
appears to be a greater vasoconstriction of the renal afferent arterioles to intrarenal
adenosine and suppressed nitric oxide bioavailability in the kidney due to endothelial
dysfunction (Komers & Anderson, 2003). Recent reports indicate that severe renal
dysfunction need not be present to create a risk of CIN in diabetic patients with measured
creatinine clearance of 100 mL/min and receiving proper hydration (Hardiek et al., 2008).

2.1.1 Prevention strategies to attenuate contrast-induced nephropathy in patients with


type 2 diabetes mellitus and coronary artery disease
Since treatment of CIN after it has occurred is ineffective, efforts to prevent or attenuate the
injury are the focus of ongoing investigations. Early diagnosis and careful long-term
treatment of patients with type 2 diabetes mellitus and coronary artery disease includes 81
mg aspirin daily, angiotensin-converting enzyme inhibitor therapy or angiotensin II
receptor blocker therapy and 3-hydroxyl-methylglutaryal coenzyme A reductase inhibitor
therapy. Ideal goals for vascular risk factor modification include blood pressure of 130/80
mmHg or better, LDL cholesterol less than 70 mg/dL and hemoglobin A1C less than 7%.
Successful accomplishment of these goals should decrease the progression of macro- and
microvascular complications. In addition, if these goals are accomplished, endothelial
function should be better. If the patient suddenly develops an acute coronary syndrome, the
ability of the kidney to have a compensatory vasodilatory response in response to contrast
medium should decrease the risk of CIN. Based on the assumption that acute renal
adenosine and/or endothelia-medicated vasoconstriction and endothelial dysfunction are
major interrelated pathogenic events in CIN, a type 2 diabetic patient meeting therapeutic
goals should be at lower risk when coronary angiography and/or coronary intervention are
required. When a patient with type 2 diabetes presents with an acute coronary syndrome,
hyperglycemia is common, and increased risk of mortality in patients has been observed
(Kosiborod & McGuire, 2010). Well-designed, large clinical outcome trials are required for
better guidance as far as how hyperglycemia in the acute situation should be managed.
Observational studies suggest that hypoglycemia should be avoided (Zoungas et al., 2010).
Guidelines now recommend a therapeutic target of glucose control at 140 to 180 mg/dL in
the critically ill patient which is a more liberal approach than prior recommendations
(O’Keefe et al., 2011). Previously recommended more aggressive glucose lowering
(including normalization of blood sugar) was not shown to have additional benefit and
appeared to be harmful because of the increased incidence of hypoglycemia.
As mechanisms for CIN in patients with type 2 diabetes mellitus are better understood,
there is a growing list of medications that may exacerbate the risk of CIN. Many of these
medications are taken by patients with cardiovascular disease and should be addressed
prior to proceeding with angiography. Medications that increase the risk of a patient
developing CIN include nonsteroidal anti-inflammatory drugs which inhibit compensatory
renal prostaglandin synthesis, diuretics that dehydrate the kidney and increase the risk of
238 Advances in the Diagnosis of Coronary Atherosclerosis

medullary ischemia, and dipyridamole that blocks the normal cellular uptake of adenosine
resulting in a greater renal adenosine – mediated vasconstrictive response.
In patients with type 2 diabetes mellitus requiring elective angiography, nonsteroidal anti-
inflammatory medicines, diuretics and dipyridamole should be held prior to the procedure.
While nonsteroidal anti-inflammatory medications are considered the cornerstone for
managing the pain of osteoarthritis and other painful conditions, recent meta-analysis
indicate significantly increased risk of myocardial infarction, stroke or death from
cardiovascular disease compared to placebo with the chronic use of these medications.
Contrary to some previous reports, the current meta-analysis found no suggestion that this
increased cardiovascular risk is specific to cyclo-oxygenase-2 inhibitors. Data were available
for naproxen, ibuprofen, diclofenac, celecoxib, etoricoxib, rofecoxib and lumiracoxib.
Overall, naproxen appeared to be the least harmful nonsteroidal anti-inflammatory
medication in terms of cardiovascular outcomes. Risks were greatest with ibuprofen,
diclofenac, etoricoxib and lumiracoxib. Thus, chronic use of nonsteroidal anti-inflammatory
medications in patients with type 2 diabetes who are at increased cardiovascular risk is
discouraged. In regard to dipyridamole, recent data suggests superiority of clopidogrel over
dipyridamole in patients who have suffered a cerebral vascular accident. Increasingly
clopidogrel rather than dipyridamole is the recommended antiplatelet agent in addition to
81 mg aspirin in patients with type 2 diabetes after transient ischemic attacks or cerebral
vascular accidents. Thus, if the patient with type 2 diabetes mellitus needs emergency
angiography, the risk of dipyridamole contributing to CIN would no longer exist.
Patients with type 2 diabetes mellitus have a significantly greater risk of macrovascular
disease, including coronary heart disease, stroke and peripheral vascular disease, in addition
to the microvascular complications, such as retinopathy, neuropathy and nephropathy.
Despite evidence of benefit for reduction of microvascular disease with glycemic control, to
date, there is no such evidence for the role of glycemic control in macrovascular event
reduction. In newly diagnosed patients with type 2 diabetes, besides initiation of diet, exercise
and weight reduction, metformin is usually the recommended initial therapy unless
contraindicated, such as renal disease (serum creatinine greater than 1.5 mg/dL in males and
greater than 1.4 mg/dL in females). Although not relevant to CIN prevention, metformin
should be temporarily discontinued at the time of or prior to use of contrast medium or major
surgical procedures and withheld for 48 hours after the procedure. Metformin should be
restarted after renal function has been reevaluated and found to be normal (Goergen et al.,
2010). While the mortality rate for patients who develop lactic acidosis can be quite high, the
actual number of people dying from metformin-induced lactic acidosis after contrast medium
exposure is quite low using these guidelines.
Cigarette smoking is the most common cause of preventable morbidity and mortality in
developed countries. In addition to it being a major risk factor for cardiovascular disease,
lung disease and cancer, cigarette smoking is now recognized as an important independent
risk factor of chronic kidney disease, including diabetic nephropathy, by increasing the rate
of transition from microalbuminuria to proteinuria and promoting the progression to end-
stage renal disease (Phisitkul et al., 2008). Furthermore, a recent cross-sectional analysis of
participants in the National Health and Nutrition Examination Survey demonstrated a
strong association between exposure to secondhand smoke and proteinuria, suggesting that
passive smokers are also at increased risk of chronic kidney disease (Obert et al., 2011).
Several clinical and experimental studies have demonstrated the role of transforming
growth factor-beta in the pathogenesis of chronic kidney disease, including diabetic
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 239

nephropathy. This cytokine is largely profibrotic and plays a significant role in diabetic
nephropathy by increasing the production of extracellular matrix proteins, including
fibronectin and collagen, in the glomerulus. It is also known that one cigarette paralyzes the
ability of the endothelium to make nitric oxide for up to 24 hours. Thus, patients with type 2
diabetes mellitus should be counseled to discontinue smoking. Emphasizing that
recommendation prior to elective angiography may decrease the incidence of CIN.
Another class of medication that warrants discussion in the long-term treatment of patients
with type 2 diabetes mellitus, not only for the long-term metabolic advantages, but also
because of the risk of CIN is beta-blocker therapy. Both nonselective and selective
traditional beta-blockers have been shown to increase insulin resistance, facilitate weight
gain of approximately 1 kilogram per six months and worsen hypertriglyceridemia by
approximately 13%. In contrast, carvedilol in hypertensive diabetic patients has been found
to have a neutral effect on insulin resistance, weight and triglycerides (Bakris et al., 2004).
This favorable metabolic profile also suggests that carvedilol is a better choice compared to
traditional beta-blockers in these high risk patients. Recent trials emphasize the class
heterogeneity that exists for beta-blockers and provide a strong basis for preferred use of
carvedilol in patients with type 2 diabetes. Carvedilol is a unique molecule that combines
the properties of a nonspecific beta-blocker and a specific alpha-1-blocker in a ratio of 2:3.
Carvedilol also possesses antioxidant properties. In hypertensive diabetic patients,
carvedilol compared to metoprolol tartrate has been shown to reduce existing
microalbuminuria and to decrease the risk of progression to microalbuminuria. These
results suggest improved endothelial function and may be related to improvement in insulin
resistance or an effect on oxidant stress in the kidney. Thus, in patients with type 2 diabetes
mellitus that require beta-blocker therapy, carvedilol has become the recommended beta-
blocker. There are also data to suggest that carvedilol compared to traditional beta-blockers
attenuates CIN. Proposed mechanisms include unopposed alpha-medicated renal
vasoconctriction causing more ischemia when on traditional beta-blockers and/or improved
endothelial function due to the antioxidant properties of carvedilol.
There are other medicines that may attenuate the risk of CIN. Calcium-channel blocking
medications attenuate both the magnitude and duration of renal vasoconstriction after
contrast medium administration (Russo et al., 1990). In patients with type 2 diabetes
mellitus and hypertension and/or angina requiring calcium-channel blocking medications,
these medications should be continued if contrast medium administration is planned.
The adenosine receptor antagonists, theophylline and aminophylline, attenuate the decrease
in GFR seen after contrast medium. Since patients with type 2 diabetes mellitus have an
increased reactivity to intrarenal adenosine, theophylline prior to contrast medium
administration warrants special consideration. The kidney responds to contrast media-
induced stress with a tubuloglomercular feedback response which is largely medicated by
adenosine when less than 100 mL of contrast medium is administered. This mechanism has
been confirmed by both animal and clinical studies. Pretreatment with long-acting
theophylline 3 mg/kg orally at least 30 minutes before contrast medium administration and
12 hours later in addition to hydration has been shown to attenuate the depression of
creatinine clearance. It is critical that theophylline be administered before contrast medium
injection, but prolonged treatment after angiography is unnecessary. Theophylline
prophylaxis is effective, safe, and inexpensive. This dose of theophylline is manyfold greater
than the minimum dose required to block renal vascular adenosine receptors and is well
below that shown to affect renal cyclic nucleotide phosphodiesterase activity (Vassallo &
240 Advances in the Diagnosis of Coronary Atherosclerosis

Lipsky, 1998). This dose of theophylline causes transient plasma theophylline levels of 7
mcg/mL which is below therapeutic levels required for treatment of asthma and unlikely to
cause serious gastrointestinal, neurological, or cardiovascular adverse effects (Cooling, 1993;
Shannon, 1999).
Adenosine receptor antagonists, theophylline and aminophylline, prophylaxis to attenuate
CIN has not been strongly adopted except in patients at high risk for CIN. Some controlled
trials have shown a significant reduction in the risk for CIN while others have shown no
reduction (Abizaid et al., 1999; Erley et al., 1999; Erley et al., 1994; Gandhi et al., 1992; Huber
et al., 2001; Huber et al., 2002; Huber et al., 2003; Kolonko et al., 1998; Shammas et al., 2001).
However, these studies have been limited by small sample size, variation in timing and
dosage of drug administration, and variation in the definition of CIN. Three meta-analyses
have dealt specifically with the clinical use of theophylline for the prevention of CIN. Each
of these gave extensive information concerning the way that the papers which were
included were chosen, exactly what factors were chosen for matching across studies and
which factors did not match. In addition, statistics were presented which addressed the
concerns about selection bias and efforts were made to adjust for non-matching factors to
the extent possible. No attempt will be made to describe each of these studies completely,
however, we will give some summary information concerning each of them. One meta-
analysis identified 10 studies in which theophylline was used for the prevention of CIN (Ix
et al., 2004). Of the ten, seven were found to fulfill all the inclusion criteria and these
constituted the final sample for the analysis. The studies included were reported between
1994 and 2003 and included a number of different protocols for the administration of the
theophylline. The final conclusion reached by these investigators was that the prophylactic
administration of theophylline appears to protect against contrast-induced declines in
kidney function. They also reported that “the protective benefits appeared robust regardless
of the study design, form or volume of contrast medium delivered, and the presence or
absence of intravenous volume expansion.” The investigators did indicate that they were
unable to determine (given the available data) if theophylline reduced the number of
patients who experienced large increases in serum creatinine concentrations or required
dialysis. Another meta-analysis identified nine studies which met the study criteria for their
protocol (Bagshaw & Ghali, 2005). Their conclusion was that the data they considered
indicated that there is promising evidence supporting the use of theophylline for the
prevention of CIN but the evidence is still not conclusive. They suggested that what was
required to answer this question was a larger, well designed clinical trial for confirmation
and assessment of risks before theophylline could be recommended routinely for the
prevention of CIN. They also noted from their analysis of these papers that such a trial
should include a hydration protocol and the routine use of low or iso-osmolar nonionic
contrast media since those are now the current standards of care. These two meta-analyses
focused primarily on theophylline and a comparison of the choices of papers from the two
studies shows that seven papers included in the initial meta-analysis were also included in
the nine chosen by the second meta-analysis. The latter authors included two papers which
were in the same search time frame as the initial meta-analysis but were not selected under
that study protocol. A third meta-analysis covered a number of drugs aimed at reducing the
incidence of CIN (Kelly et al., 2008). The primary drug considered was N-acetylcysteine
with theophylline being more secondary to the study. Only six of the studies considered
dealt primarily with theophylline. Of the six considered, five were part of the set of studies
considered by the second meta-analysis while four of them coincided with those used in the
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 241

initial meta-analysis. The sixth paper which was unique to this analysis appeared in 2006
which was out of the range of years covered by the other two papers. One of the six papers
common to both was not included by the initial meta-analysis. The conclusion of this paper
with respect to the efficacy of theophylline was that it produced a large risk reduction but
did not achieve statistical significance. Of these three meta-analyses, the first two are most
comparable. Both reach the same conclusion. Each of these papers used slightly different
statistical methods. Both contained a thorough discussion of the selection criteria used to
choose the papers included and measures to check for selection bias. The general
conclusions were favorable with the hypothesis that theophylline is protective against CIN.
Unfortunately, neither group of authors was willing to report a definitive answer. There are
some areas where the studies under review by these authors differed. One was in the area
of how the theophylline was administered: as a fixed dose regardless of body weight or as a
dose adjusted per kilogram of body weight. The broad conclusion reached is that a large,
carefully designed clinical trial needs to be conducted to reach a final conclusion. The trial
should be stratified according to ancillary risk factors such as age and diabetes. In addition,
the effect of the dosing protocol and hydration methods needs to be considered. Finally,
since it is known that certain beverages like coffee and tea also block adenosine vascular
receptors, a study testing the benefit of theophylline should exclude other xanthine
exposure during the time before contrast medium administration and for the 48 hours after
while the subject is being followed.
Studies suggest two mechanisms by which intrarenal adenosine contributes to CIN. Early
depression of renal function after contrast medium exposure is caused by A1 receptor-
mediated renal afferent arteriolar vasoconstriction and A2 receptor-mediated efferent
arteriolar vasodilatation resulting in decreased glomerular perfusion pressure. Later
depression of renal function after contrast medium exposure is caused by oxygen-free
radical production, in part, due to intrarenal adenosine catabolism to xanthine.
Pretreatment with theophylline may attenuate CIN by maintaining GFR such that contrast
medium is cleared from the kidney more rapidly, thus, decreasing direct cytotoxic effects
from contrast medium and decreasing the potential for intrarenal adenosine-mediated
oxygen-free radical production. Pretreatment with allopurinol has also been shown to
attenuate CIN. To be effective, it is crucial to administer allopurinol at least 24 hours prior to
exposure to contrast medium. Pretreatment allows time for production of oxypurinol, the
chief metabolite of allopurinol, which is a much more potent xanthine oxidase inhibitor.
Allopurinol administered in this fashion was renally protective. Decreasing renal xanthine
oxidase decreases the catabolism of intrarenal adenosine to xanthine resulting in decreased
oxygen-free radical production and less renal tubular cell injury.
Patients with type 2 diabetes mellitus are usually on angiotensin-converting enzyme
inhibitors, angiotensin II receptor blockers and/or direct renin inhibitor to decrease the
progression of renal disease. Whether these agents need to be held or should be continued
when contrast medium is administered requires clinical judgment. Intrarenal angiotensin II
enhances adenosine-mediated vasoconstriction so theoretically blockade of the renin-
angiotensin system should attenuate the risk of CIN. In support of this concept,
preprocedure angiotensin-converting enzyme inhibitor use has been shown to lower the risk
for CIN in patients with chronic renal disease (Dangas et al., 2005). Another clinical trial
showed that periprocedural captopril, compared with an untreated control group, reduced
the risk of CIN in patients with type 2 diabetes mellitus (Gupta et al, 1999). However, if an
unstable patient has low cardiac output or hypotension, renin-angiotensin system blockade
242 Advances in the Diagnosis of Coronary Atherosclerosis

is known to worsen glomerular perfusion (i.e. intrarenal angiotensin II is needed). In such


patients, angiotensin-converting inhibitor therapy has increased the risk of CIN (Cirit et al.,
2006; Holscher et al., 2008; Louis et al., 1996; Rosenstock et al., 2008). Given the long-term
benefit of angiotensin-converting enzyme inhibitor and angiotensin II receptor antagonists,
most believe these medications should be continued in patients with diabetes mellitus and
chronic kidney disease requiring contrast medium administration unless the patient is
hemodynamically unstable. However, if angiotensin-converting enzyme inhibitors or
angiotensin II receptor blockers are held before contrast medium administration, they
should be restarted when renal function is considered stable.
Recent research suggests that 3-hydroxy-methylglutaryl coenzyme A reductase inhibitors,
or statins, may reduce the risk of CIN because they have beneficial effects on endothelial
function, maintain nitric oxide production and reduce oxidative stress (Adel et al., 2010).
Recent retrospective reviews of patients with renal impairment undergoing angiography
suggests that the risk of CIN was lower in patients in whom a statin was initiated before the
procedure (Khanal et al., 2005). These findings reinforce the rationale for the introduction of
statin therapy before undergoing diagnostic or interventional coronary angiography,
particularly in patients with type 2 diabetes mellitus (Attallah et al., 2004).
Hypomagnesemia, serum magnesium level less than 2 mEq/L, may also be a correctable
risk factor for CIN in patients with renal dysfunction. Patients with type 2 diabetes mellitus
frequently become hypomagnesemic due to poor control with glycosuria, the development
of an acquired renal tubular defect and from chronic diuretic therapy. All three conditions
decrease the renal tubular reabsorption of magnesium. Detecting hypomagnesemia and
correcting it prior to angiography or coronary intervention may be indicated. The renal
protective effect of magnesium is likely multifactorial. Besides its role as an antioxidant and
as a coenzyme for compensatory sodium-potassium adenosine triphosphatase, magnesium
has calcium channel blocking properties.
The universally accepted prevention strategy for CIN is adequate intravenous volume
expansion. Extracellular volume expansion plays a well established role in reducing the risk
of CIN although few trials have directly addressed the ideal protocol. Intravenous volume
expansion before and after the administration of contrast medium appears to be more
effective than either bolus volume expansion during the procedure or removal of
restrictions on oral fluid intake. Isotonic saline has been found to be better than 0.45% saline
and is given intravenously before and after administration of contrast medium for a total of
24 hours. An additional strategy that is under study is for patients to receive prophylactic
volume expansion with isotonic sodium bicarbonate solution (Merten et al., 2004). It is
thought that an alkaline environment decreases oxygen-free radical formation in the renal
tubule. The findings of several recent trials and a meta-analysis indicate that volume
expansion with sodium bicarbonate is more effective than volume expansion with saline.
However, recent reports argue against a clear benefit for sodium bicarbonate volume
expansion but examination of the various trials suggests that the dose given to accomplish
an alkaline environment in the renal tubule is important. Isotonic sodium bicarbonate
solution, administered at 3 mL/kg/h for one hour prior to angiography and at 1 mL/kg/h
for, at least, six hours afterward accomplishes that goal. A major robust, well-designed trial
comparing volume expansion with sodium bicarbonate versus normal saline is ongoing. At
this time, the CIN consensus working panel concludes that no adjunctive medical
pretreatment has been validated as effective for preventing CIN. Use of furosemide,
mannitol or an endothelin receptor antagonist is potentially detrimental. Felodipine,
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 243

dopamine, atrial natriuretic peptide and L-arginine have not been shown to be effective.
Potentially beneficial strategies for attenuating CIN include theophylline, allopurinol,
ascorbic acid, oral and intravenous N–acetylcysteine, hemodialysis of contrast medium and
venous capture of contrast medium.
In regard to pretreatment approaches to limit oxygen-free radical injury following contrast
medium, four pharmacologic approaches warrant further study with adequately powered,
appropriately designed, randomized trials of high-risk diabetic patients, perhaps combined
with theophylline pretreatment. One of these is isotonic sodium bicarbonate solution with
the length of post-procedure treatment determined by the volume of contrast medium
administered. Maintaining an alkaline environment should decrease oxygen-free radical
formation in the renal tubule. As discussed above, one source of oxygen-free radical
production after contrast medium is the catabolism of intrarenal adenosine to xanthine.
Allopurinol, a xanthine oxidase inhibitor, 4 mg/kg orally daily starting 24 hours before
administration of contrast medium has shown benefit. This dose of allopurinol, if given 24
hours before elective procedures, is metabolized into oxypurinol which is a more effective
xanthine oxidase inhibitor than allopurinol. This 24-hour pretreatment approach has been
shown to attenuate the fall in GFR after contrast medium exposure. Besides limiting oxygen-
free radical formation, allopurinol may also protect the kidney from contrast medium
exposure by its ability to inhibit adenine nucleotide degradation (thus, preservation of
adenine nucleotide is required for recovery from renal injury). Allopurinol has also been
found to markedly decrease the vasodilatation response to intravenous adenosine in the
renal vasculature. Less adenosine-mediated efferent renal arteriolar vasodilatation would
preserve glomerular perfusion pressure. In view of the possible role of oxidase stress and
oxygen-free radical generation in CIN, ascorbic acid as an antioxidant warrants further
study (Spargias et al., 2004). N-acetylcysteine, as an antioxidant and renal vasodilator, has
not been shown to be consistently effective when given orally (Solomon, 2009). Perhaps a
larger oral dose is needed or intravenous administration may be required. Allopurinol,
ascorbic acid and N-acetylcysteine, if proven beneficial, require pretreatment and would be
less helpful in diabetic patients requiring emergency diagnostic and/or interventional
contrast medium usage.
To decrease the risk of CIN requires careful patient screening and selection, adequate
patient hydration, limiting the volume of contrast medium administered and choosing a
safe, non-ionic, low osmolar contrast agent. Based on comparisons of contrast media in
proximal renal tubular cell culture and in recent robust head-to-head prospective clinical
trials in high risk patients, however, iso-osmolar iodixanol and low-osmolar iopamidol are
comparable and appear to be the contrast agents of choice to reduce renal risk for CIN.
The direct cellular toxicities of commonly used contrast medium have been compared in
renal proximal tubular cells in culture (Hardiek et al., 2001). All contrast media, whether
high-osmolar or iso-osmolar, ionic or non-ionic, stress renal proximal tubular cells. When
renal proximal tubular cells in culture are exposed to contrast media, they respond by
increasing extracellular adenosine concentrations because of the depletion of adenosine
triphosphate due to osmotic load and the large size of contrast medium molecules. The renal
toxicity from the direct effects of contrast medium is reversible, as has been shown in vitro in
studies in which renal tubular cells respond to contrast media exposure by decreasing the
activity of mitochondrial enzymes without altering viability. While all contrast medium may
reduce mitochondrial enzyme activity, differences among agents are seen with iopamidol
and iodixanol being the least toxic and the two ionic contrast media, ioxaglate and
244 Advances in the Diagnosis of Coronary Atherosclerosis

diatrizoate, being the most toxic. The lesser the depression of mitochondrial function, the
more rapidly renal proximal tubular cells can recover after contrast medium has been
eliminated from the kidney suggesting that iopamidol and iodixanol are the least
nephrotoxic molecules when studied in cell culture.
Earlier clinical trials comparing contrast medium risk in patients with type 2 diabetes for
CIN had limitations that prevented valid conclusions: the studies were not blinded, the
number of patients was small, the timing of outcome assessment was unclear, or the patients
received contrast intra-arterially and/or intravenously. With proper hydration and
identification of high-risk patients, the incidence of CIN after intra-arterial contrast medium
administration is about 10%. More recent trials in high-risk diabetic patients given contrast
medium intravenously suggest that the incidence of CIN is about 5%. More recent trials
have been head-to-head, robust and prospective in high-risk patients with chronic renal
failure (eGFR 59-20 mL/min) with intra-arterial contrast medium administration. Based on
animal studies, concern about osmotoxicity in the pathogenesis of CIN emerged which led
to the development of low-osmolar, and, later, iso-osmolar contrast medium. Of interest, in
human prospective trials and meta-analysis, no specifically significant differences in
nephrotoxicity between high-osmolar (1,400-2,000 mOsm/kg) and non-ionic low-osmolar
(600-800 mOsm/kg) contrast have been found in patients with normal renal function
(Solomon, 2005). Clinical trials published in the 1990s, however, showed that use of high-
osmolar ionic monomer diatrizoate in patients with chronic renal disease who were
undergoing coronary angiography had a higher incidence of CIN than that associated with
non-ionic low-osmolar contrast medium. In general, lower osmolar contrast medium has
now replaced diatrizoate for routine clinical use. It is less clear whether there are
appreciable differences among various non-ionic low-osmolar contrast medium regarding
the incidence of CIN. Because individual contrast medium has specific effects on renal
tubular cells, head-to-head studies are required to compare the safer contrast medium in at
risk patients undergoing angiography. With the development of the non-ionic iso-osmolar
dimer, iodixanol, subsequent studies have evaluated whether a further reduction in
osmolality would result in still more protection against CIN. In the first of these
comparative studies ((Nephrotoxicity of High-Risk Patient Study of Iso-osmolar and Low-
Osmolar Non-ionic Contrast Medium (NEPHRIC)) 129 patients with diabetes with chronic
renal failure (baseline eGFR of 48 mL/min) and serum creatinine levels 1.5-3.5 mg/dL were
randomized to the low-osmolar contrast medium iohexol or the iso-osmolar contrast
medium, iodixanol (Aspelin et al., 2003). A higher rate of CIN defined as a serum creatinine
rise of greater than 0.5 mg/dL after angiography was noted after iohexol (26.2%) than after
iodixanol (3.1%).
Since the publication of NEPHRIC, subsequent prospective randomized trials involving
high-risk patients with renal insufficiency and diabetes and the intra-arterial administration
of contrast medium have not consistently found a lower incidence of CIN associated with
iso-osmolar contrast medium. In general, a benefit favoring the non-ionic iso-osmolar dimer,
iodixanol, is seen compared with the non-ionic low-osmolar monomer iohexol or the ionic
low-osmolar dimer, ioxaglate. In contrast, when comparing non-ionic low-osmolar
monomers, iopamidol or ioversol, in high-risk patients with iso-osmolar iodixanol, no
additional protective effect is seen. There have been no head-to-head comparisons of iohexol
with any other non-ionic, low-osmolar contrast agents in high-risk patients undergoing
intra-arterial administration of contrast. Based on a recent systematic review of
angiographic contrast medium in high-risk patients, however, the likely explanation of the
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 245

NEPHRIC findings is that low-osmolar, iohexol, appears to be more nephrotoxic than other
low-osmolar contrast media, such as iopamidol and ioversol. Furthermore, a systematic
review of angiographic contrast media in high-risk patients found no statistically significant
difference in the risk of CIN between low-osmolar iopamidol and iso-osmolar iodixanol
(Solomon, 2005). These comparisons in low-osmolar and iso-osmolar contrast medium
administered intra-arterially in high-risk patients suggest that each molecules safety must
be based on robust clinical trials. These data also suggest that physical and/or chemical
properties of each molecule, other than osmolarity, are implicated in the pathogenesis of
CIN. Given this incidence of CIN, prospective randomized trials need larger patient
populations to be properly powered to be certain of the conclusions. The recently published
Cardiac Angiography in Renally-Impaired Patients (CARE) study fits these criteria by being
a multicenter, double-blind, randomized study designed to prospectively compare the
incidence of CIN after intra-arterial administration of low-osmolar iopamidol or iso-osmolar
iodixanol in 414 patients with moderate to severe chronic kidney disease (eGFR of 20-59
mL/min/1.73 m2) who underwent coronary angiography or percutaneous coronary
intervention (Solomon et al., 2007). All patients received intravenous sodium bicarbonate
prophylaxis. The primary endpoint was a post-dose serum creatinine increase of greater
than 0.5 mL/dL over baseline. The renal effects of these two agents were comparable with
no significant difference in occurrence of CIN and no significant difference in occurrence of
CIN in the subgroup of chronic kidney disease patients with diabetes. More recently, a
multicenter, randomized double-blind trial comparing the renal effects of non-ionic, iso-
osmolar iodixanol versus non-ionic, low-osmolar iopamidol in 526 patients with impaired
baseline renal function and diabetes undergoing diagnostic and/or therapeutic coronary
angiography procedures was completed (Laskey et al., 2009). This trial showed overall rate
of CIN in patients with chronic kidney disease and diabetes undergoing coronary
angiographic procedures was 10.5%. There was no significant difference between iodixanol
and iopamidol in either peak increase in serum creatinine or risk of CIN.
The renal effects of iodixanol and iopamidol are comparable in high-risk diabetic patients
requiring contrast medium for diagnostic studies or intervention. The non-renal differences
between these contrast agents should be considered when selecting an agent and when
obtaining informed patient consent, however. Using a contrast medium with a higher iodine
concentration per milliliter, such as iopamidol which has 370 mg/mL compared to iodixanol
which has 320 mg/mL iodine content, may allow for a smaller volume to be used
intravenously without sacrificing image quality. The higher iodine concentration of
iopamidol may make complex plaques easier to appreciate during coronary angiography
and intervention. Other considerations are that iodixanol is threefold more likely to cause
contrast-induced delayed skin reactions compared to iopamidol. After percutaneous
coronary intervention with bare-metal or drug-eluting stent placement, cardiac patients
require antiplatelet therapy with clopidogrel, which may cause a rash within 5 to 7 days of
treatment initiation, a timeline similar to that for delayed contrast-induced skin reactions. In
clinical trials with clopidogrel, skin reactions are noted in 4.2% of patients. The choice of a
contrast molecule, such as iopamidol, that is less likely to cause a delayed contrast-induced
skin reaction makes decisions regarding the discontinuation of clopidogrel less frequent and
more definitive. Finally, iopamidol is less expensive than iodixanol.
Gadolinium-based contrast agents were introduced, partly, because of the discovery that
iodine-based contrast medium could cause CIN. However, recent reports suggest that
gadolinium-based agents may also be nephrotoxic. Furthermore, after exposure to
246 Advances in the Diagnosis of Coronary Atherosclerosis

gadolinium-based contrast, some patients with renal insufficiency have developed


nephrogenic systemic sclerosis with scleroderma-like changes in the skin, connective tissues
and other organs, which has sometimes been fatal (Grobner & Prischl, 2007). In diabetic
patients with chronic renal failure, the use of any contrast agent should be avoided, if
possible, but, if required, small volumes of a contrast agent, such as iodixanol or iopamidol
with proper hydration appears to be the safest clinical management to reduce renal and
long-term patient risk.
Ongoing investigation is searching for even more sensitive markers that may predict the
development of CIN after percutaneous coronary interventions in patients with normal
serum creatinine values (Kato et al., 2008). Future studies should include cystatin C which is
an alternative serum measurement of kidney function that approximates direct measures of
GFR more precisely than creatinine because its serum concentrations are independent of
muscle mass and do not appear to be affected by age or sex. Cystatin C may identify a
preclinical risk of kidney dysfunction after contrast medium that is not detected by serum
creatinine or eGFR (Perkins et al., 2005). More recently, the value of neutrophil gelatinase-
associated lipocalin (NGAL) has been highlighted as a novel biomarker for the early
detection of acute renal failure (Ling et al., 2008). NGAL, a member of the lipocalin family,
was originally isolated from the supernatant of activated human neutrophils, but it is also
expressed at a low level in human tissues, including the kidney. Because of its small
molecular size and resistance to degradation, NGAL is readily excreted and detected in
urine. Thus, NGAL is highly accumulated in human kidney cortical tubules, blood and
urine after renal injuries. NGAL might represent an early, sensitive and noninvasive urinary
marker for the detection of CIN. Initial studies of NGAL after percutaneous coronary
intervention showed an earlier increase in serum than in urine probably because NGAL was
released into the circulation secondary to inflammatory activation of neutrophils initiated by
percutaneous coronary intervention. Moreover, since NGAL is increased in atherosclerotic
plaques, it might also be released in the circulation during the PCI. It has been found that
predictors of serum NGAL 2 hours after PCI were serum creatinine, cystatin C, length of the
percutaneous coronary intervention and the presence of diabetes. These findings have
important implications for the clinical management of diabetic patients undergoing PCI.
Since patients after PCI are often discharged the next day and CIN often occurs subsequent
to discharge, NGAL needs to be investigated as a potential early marker for nephrotoxicity.
Further studies in type 2 diabetes mellitus patients with chronic renal failure undergoing
contrast medium administration are warranted to assess the use of NGAL with respect to
earlier detection of CIN so that patients can be advised properly and medicines, such as
metformin and angiotensin-converting enzyme inhibitors or angiotensin II receptor
blockers, can be restarted appropriately.
In patients undergoing cardiac catheterization, cystatin C may be useful in predicting before
catheterization the risk of CIN. In contrast, urinary human liver-type fatty acid-binding
protein (L-FABP) consistently appears to be the only marker for detecting transient renal
tubular damage in patients with moderate renal disease. Thus, urinary L-FABP appears to
be a useful marker of renal tubular damage caused by contrast medium. Other investigators
have found that urinary intraleukin-18 (IL-18) is associated with the later cardiac outcomes
in patients with CIN after coronary angiography. CIN, a form of acute kidney injury, has
received increasing attention the past few years because of the recognition that CIN is
associated with long-term adverse events. The increased incidence of adverse events after
CIN is derived primarily from retrospective analysis of large data bases or observational
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 247

studies of patients who have undergone coronary angiography and/or percutaneous


coronary intervention. A cause-and-effect relationship cannot be determined from such
data. Patients with an increased burden of cardiovascular risk factors before contrast
medium exposure may be more likely to develop CIN and independent of the occurrence of
CIN may have more long-term adverse events (Shlipak et al., 2005). Alternatively, the
occurrence of CIN may in some as yet undefined manner alter the future likelihood of
adverse events (i.e., CIN triggers a pathophysiologic pathway that leads to adverse events).
The Cardiac Angiography in REnally-impaired patients (CARE) study has allowed the
opportunity to explore the association of CIN with long-term adverse events (Solomon et al.,
2009). Patients were followed for the year after the trial, and adverse events were collected.
The rate of long-term adverse events was higher in individuals who developed CIN. After
adjustment for baseline comorbidities and risk factors, the adjusted incident ratio for
adverse events was twice as high in those with CIN compared to the patients who did not
develop CIN. It is also noted that randomization to iopamidol reduced the incidence of CIN
and adverse events compared to iodixanol. Parallel decrease of incidence of CIN and
adverse events in the iopamidol arm of this randomized trial supports a causal role for CIN
in late adverse events in diabetic patients undergoing coronary angiography and/or
coronary intervention. Why a transient decrease in renal function results in long-term
increased risk of cerebrovascular accident, myocardial infarction and death is an area of
intense study. Some investigators report that traditional cardiovascular risk factors have
greater associations with cardiovascular mortality than novel risk factors. Thus, in patients
that develop CIN, continued treatment of hypertension, hyperlipidemia and diabetes is
recommended. Since multiple studies have demonstrated the important role of nitric oxide
in endothelial physiology and the pathogenesis of vascular disease, ongoing studies are
examining endogenous nitric oxide synthase inhibitors, such as the amino acid asymmetric
dimethylarginine, in the pathogenesis of late adverse events (Chirinos et al., 2008).
Additional studies are required to assess the prognostic value of measuring for this
endogenous nitric oxide synthase inhibitor and assessing whether interventions limiting its
effect will benefit patients at risk for future cardiovascular events.
2.1.1.1 Risks and benefits of prevention strategies to attenuate contrast-induced
nephropathy in patients with type 2 diabetes mellitus and coronary artery disease
The risks of a short course of theophylline, either orally or intravenously, is low. If a patient
is on theophylline, one cannot give adenosine as a coronary vasodilator as part of a flow
wire analysis to assess the functional significance of a coronary stenotic lesion (Casella et al.,
2003; Yoon et al., 2009). However, intravascular ultrasound can be used as the guide for
functional significance, and, thus, pretreatment with theophylline to attenuate CIN may be
more important than losing this one technique of assessing functional coronary stenosis. Of
the various potential strategies for attenuating CIN, theophylline can be administered orally
or intravenously within 30 minutes of the procedure and be beneficial. Antioxidant
strategies, such as allopurinol, ascorbic acid, or N-acetylcysteine require pretreatment and
will be less helpful in patients requiring emergency diagnostic and/or interventional
contrast medium usage.
Volume expansion with normal saline or sodium bicarbonate is the universally agreed upon
strategy to attenuate CIN. Bicarbonate therapy was initially explored because the generation
of oxygen-free radicals is pH dependent through the Haber-Weiss reaction (Merten et al.,
2004). Alkalinization of the urinary space is achieved very quickly with intravenous infusion
248 Advances in the Diagnosis of Coronary Atherosclerosis

of sodium bicarbonate because normally there is little bicarbonate in the urine. Even a small
increase in serum bicarbonate of 1 to 2 mEq/L will result in the ‘dumping’ of bicarbonate
into the urine in most patients (Solomon, 2009). Such a change in serum is easily obtained
with the infusion rate recommended. Sodium bicarbonate therapy is readily available,
inexpensive, and safe. The question is whether it is efficacious for prevention of CIN. Of
particular clinical interest from recent meta-analysis was the finding that sodium
bicarbonate therapy is most effective in patients who experienced urgent or emergency
contrast medium exposure (Meier et al., 2009). Presumably this selects a group of patients
who are less likely to receive any other form of prophylaxis for CIN. This is of great
potential importance for the emergency room and cardiac catheterization laboratory.
Sodium bicarbonate therapy was also most effective in those receiving low osmolality
contrast media compared with iso-osmolality contrast media. Low osmolality contrast is
increasingly chosen because of its safety, lower costs, and higher iodine content. It has also
been found that the addition of N-acetylcysteine to fluid expansion with sodium bicarbonate
showed no additional reduction in the rate of CIN after the intra-arterial administration of
iopamidol or iodixanol to high-risk patients with type 2 diabetes mellitus and chronic
kidney disease (Staniloae et al., 2009).
CIN needs to be redefined using markers of kidney injury that are sensitive, specific, and
predictive of adverse outcomes (Solomon, 2009). This will enable investigators to better
address the question of how to attenuate this condition in the future. The most important
question to be answered is whether prevention of kidney injury results in a change in short-
and long-term adverse outcomes. Using iopamidol has been associated with a reduction in
long-term adverse events (Solomon et al., 2007). The recent meta-analyses found that despite
a reduction in the incidence of CIN, sodium bicarbonate therapy had no benefit on the need
for dialysis or mortality (Meier et al., 2009). No matter how available, inexpensive, and safe
a potential preventive therapy, to find an important role in clinical therapeutics, it must
improve the ‘downstream’ adverse outcomes, an as yet elusive goal for the prevention and
treatment of CIN (Solomon, 2009).

3. Conclusions
In summary, CIN remains an important clinical challenge in patients with type 2 diabetes
mellitus with coronary artery disease. Reducing the incidence of CIN requires recognizing
high risk patients, such as patients with type 2 diabetes mellitus. Long-term risk factor
modification should improve endothelial function so a more normal renal compensatory
response will occur should contrast medium administration be required. Lowest volume of
iopamidol needed would appear to be the contrast agent of choice. In patients with type 2
diabetes mellitus, pretreatment with theophylline to block adenosine-mediated renal
vasoconstriction combined with volume expansion with sodium bicarbonate to limit
oxygen-free radical generation is a rational approach based on existing data. Appropriately
designed robust randomized clinical trials combining pharmacologic approaches aimed at
both the renal vasoconstriction and the oxygen-free radical generation are needed to confirm
that this is the best preventive strategy in patients with type 2 diabetes mellitus.

4. Acknowledgement
Special thanks to Marcey R. Ervin and Sandy Sledge for preparation of the manuscript.
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 249

5. References
Abizaid, A. S., C. E. Clark, G. S. Mintz, S. Dosa, J. J. Popma, A. D. Pichard, L. F. Satler, M.
Harvey, K. M. Kent, & M. B. Leon. (1999). Effects of dopamine and aminophylline
on contrast-induced acute renal failure after coronary angioplasty in patients with
preexisting renal insufficiency. American Journal of Cardiology 83 (2):pp. ISSN 260-
263, A265, 0002-9149.
Adel, A., Z. Abdel-Salam, & W. Nammas. (2010). Low-dose statin therapy improves
endothelial function in type 2 diabetic patients with normal serum total cholesterol:
a randomized placebo-controlled study. J Clin Hypertens (Greenwich) 12 (10):pp. 820-
825, ISSN 1751-7176.
Arakawa, K., H. Suzuki, M. Naitoh, A. Matsumoto, K. Hayashi, H. Matsuda, A. Ichihara, E.
Kubota, & T. Saruta. (1996). Role of adenosine in the renal responses to contrast
medium. Kidney International 49 (5):pp. 1199-1206, ISSN 0085-2538.
Aspelin, P., P. Aubry, S. G. Fransson, R. Strasser, R. Willenbrock, & K. J. Berg. (2003).
Nephrotoxic effects in high-risk patients undergoing angiography. New England
Journal of Medicine 348 (6):pp. 491-499, ISSN 1533-4406.
Attallah, N., L. Yassine, J. Musial, J. Yee, & K. Fisher. (2004). The potential role of statins in
contrast nephropathy. Clinical Nephrology 62 (4):pp. 273-278, ISSN 0301-0430
Bagshaw, S. M., & W. A. Ghali. (2005). Theophylline for prevention of contrast-induced
nephropathy: a systematic review and meta-analysis. Archives of Internal Medicine
165 (10):pp. 1087-1093, ISSN 0003-9926.
Bakris, G. L., V. Fonseca, R. E. Katholi, J. B. McGill, F. H. Messerli, R. A. Phillips, P. Raskin, J.
T. Wright, Jr., R. Oakes, M. A. Lukas, K. M. Anderson, & D. S. Bell. (2004).
Metabolic effects of carvedilol vs metoprolol in patients with type 2 diabetes
mellitus and hypertension: a randomized controlled trial. JAMA 292 (18):pp. 2227-
2236, ISSN 1538-3598.
Casella, G., J. Rieber, T. M. Schiele, H. U. Stempfle, U. Siebert, M. Leibig, K. Theisen, U.
Buchmeier, & V. Klauss. (2003). A randomized comparison of 4 doses of
intracoronary adenosine in the assessment of fractional flow reserve. Zeitschrift fur
Kardiologie 92 (8):pp. 627-632, ISSN 0300-5860.
Chirinos, J. A., R. David, J. A. Bralley, H. Zea-Diaz, E. Munoz-Atahualpa, F. Corrales-
Medina, C. Cuba-Bustinza, J. Chirinos-Pacheco, & J. Medina-Lezama. (2008).
Endogenous nitric oxide synthase inhibitors, arterial hemodynamics, and
subclinical vascular disease: the PREVENCION Study. Hypertension 52 (6):pp. 1051-
1059, ISSN 1524-4563.
Cirit, M., O. Toprak, M. Yesil, S. Bayata, N. Postaci, L. Pupim, & E. Esi. (2006). Angiotensin-
converting enzyme inhibitors as a risk factor for contrast-induced nephropathy.
Nephron Clin Pract 104 (1):pp. c20-27, ISSN 1660-2110.
Clark, B. A., D. Kim, & F. H. Epstein. (1997). Endothelin and atrial natriuretic peptide levels
following radiocontrast exposure in humans. American Journal of Kidney Diseases 30
(1):pp. 82-86, ISSN 0272-6386.
Cooling, D. S. (1993). Theophylline toxicity. Journal of Emergency Medicine 11 (4):pp. 415-425,
ISSN 0736-4679.
250 Advances in the Diagnosis of Coronary Atherosclerosis

Dangas, G., I. Iakovou, E. Nikolsky, E. D. Aymong, G. S. Mintz, N. N. Kipshidze, A. J.


Lansky, I. Moussa, G. W. Stone, J. W. Moses, M. B. Leon, & R. Mehran. (2005).
Contrast-induced nephropathy after percutaneous coronary interventions in
relation to chronic kidney disease and hemodynamic variables. American Journal of
Cardiology 95 (1):pp. 13-19, ISSN 0002-9149.
Erley, C. M., S. H. Duda, D. Rehfuss, B. Scholtes, J. Bock, C. Muller, H. Osswald, & T. Risler.
(1999). Prevention of radiocontrast-media-induced nephropathy in patients with
pre-existing renal insufficiency by hydration in combination with the adenosine
antagonist theophylline. Nephrology, Dialysis, Transplantation 14 (5):pp. 1146-1149,
ISSN 0931-0509.
Erley, C. M., S. H. Duda, S. Schlepckow, J. Koehler, P. E. Huppert, W. L. Strohmaier, A.
Bohle, T. Risler, & H. Osswald. (1994). Adenosine antagonist theophylline prevents
the reduction of glomerular filtration rate after contrast media application. Kidney
International 45 (5):pp. 1425-1431, ISSN 0085-2538.
Gandhi, M. R., P. Brown, C. A. Romanowski, S. K. Morcos, S. Campbell, A. M. el Nahas, & T.
A. Gray. (1992). The use of theophylline, an adenosine antagonist in the prevention
of contrast media induced nephrotoxicity. British Journal of Radiology 65 (777):pp.
838, ISSN 0007-1285.
Goergen, S. K., G. Rumbold, G. Compton, & C. Harris. (2010). Systematic review of current
guidelines, and their evidence base, on risk of lactic acidosis after administration of
contrast medium for patients receiving metformin. Radiology 254 (1):pp. 261-269,
ISSN 1527-1315.
Grobner, T., & F. C. Prischl. (2007). Gadolinium and nephrogenic systemic fibrosis. Kidney
International 72 (3):pp. 260-264, ISSN 0085-2538.
Gupta, R. K., A. Kapoor, S. Tewari, N. Sinha, & R. K. Sharma. (1999). Captopril for
prevention of contrast-induced nephropathy in diabetic patients: a randomised
study. Indian Heart Journal 51 (5):pp. 521-526, ISSN 0019-4832.
Hardiek, K., R. E. Katholi, V. Ramkumar, & C. Deitrick. (2001). Proximal tubule cell response
to radiographic contrast media. Am J Physiol Renal Physiol 280 (1):pp. F61-70, ISSN
0363-6127.
Hardiek, K. J., R. E. Katholi, R. S. Robbs, & C. E. Katholi. (2008). Renal effects of contrast
media in diabetic patients undergoing diagnostic or interventional coronary
angiography. Journal of Diabetes and Its Complications 22 (3):pp. 171-177, ISSN 1056-
8727.
Holscher, B., C. Heitmeyer, M. Fobker, G. Breithardt, R. M. Schaefer, & H. Reinecke. (2008).
Predictors for contrast media-induced nephropathy and long-term survival:
prospectively assessed data from the randomized controlled Dialysis-Versus-
Diuresis (DVD) trial. Canadian Journal of Cardiology 24 (11):pp. 845-850, ISSN 1916-
7075.
Huber, W., B. Jeschke, M. Page, W. Weiss, H. Salmhofer, U. Schweigart, K. Ilgmann, J.
Reichenberger, B. Neu, & M. Classen. (2001). Reduced incidence of radiocontrast-
induced nephropathy in ICU patients under theophylline prophylaxis: a
prospective comparison to series of patients at similar risk. Intensive Care Medicine
27 (7):pp. 1200-1209, ISSN 0342-4642.
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 251

Huber, W., C. Schipek, K. Ilgmann, M. Page, M. Hennig, A. Wacker, U. Schweigart, L.


Lutilsky, C. Valina, M. Seyfarth, A. Schomig, & M. Classen. (2003). Effectiveness of
theophylline prophylaxis of renal impairment after coronary angiography in
patients with chronic renal insufficiency. American Journal of Cardiology 91 (10):pp.
1157-1162, ISSN 0002-9149.
Ix, J. H., C. E. McCulloch, & G. M. Chertow. (2004). Theophylline for the prevention of
radiocontrast nephropathy: a meta-analysis. Nephrology, Dialysis, Transplantation 19
(11):pp. 2747-2753, ISSN 0931-0509.
Kapoor, A., S. Kumar, S. Gulati, S. Gambhir, R. S. Sethi, & N. Sinha. (2002). The role of
theophylline in contrast-induced nephropathy: a case-control study. Nephrology,
Dialysis, Transplantation 17 (11):pp. 1936-1941, ISSN 0931-0509.
Kato, K., N. Sato, T. Yamamoto, Y. K. Iwasaki, K. Tanaka, & K. Mizuno. (2008). Valuable
markers for contrast-induced nephropathy in patients undergoing cardiac
catheterization. Circ J 72 (9):pp. 1499-1505, ISSN 1346-9843.
Kelly, A. M., B. Dwamena, P. Cronin, S. J. Bernstein, & R. C. Carlos. (2008). Meta-analysis:
effectiveness of drugs for preventing contrast-induced nephropathy. Annals of
Internal Medicine 148 (4):pp. 284-294, ISSN 1539-3704.
Khanal, S., N. Attallah, D. E. Smith, E. Kline-Rogers, D. Share, M. J. O'Donnell, & M.
Moscucci. (2005). Statin therapy reduces contrast-induced nephropathy: an analysis
of contemporary percutaneous interventions. American Journal of Medicine 118
(8):pp. 843-849, ISSN 0002-9343.
Kolonko, A., A. Wiecek, & F. Kokot. (1998). The nonselective adenosine antagonist
theophylline does prevent renal dysfunction induced by radiographic contrast
agents. J Nephrol 11 (3):pp. 151-156, ISSN 1121-8428.
Komers, R., & S. Anderson. (2003). Paradoxes of nitric oxide in the diabetic kidney. Am J
Physiol Renal Physiol 284 (6):pp. F1121-1137, ISSN 0363-6127.
Kosiborod, M., & D. K. McGuire. (2010). Glucose-lowering targets for patients with
cardiovascular disease: focus on inpatient management of patients with acute
coronary syndromes. Circulation 122 (25):pp. 2736-2744, ISSN 1524-4539.
Laskey, W., P. Aspelin, C. Davidson, M. Rudnick, P. Aubry, S. Kumar, F. Gietzen, & M.
Wiemer. (2009). Nephrotoxicity of iodixanol versus iopamidol in patients with
chronic kidney disease and diabetes mellitus undergoing coronary angiographic
procedures. American Heart Journal 158 (5):pp. 822-828 e823, ISSN 1097-6744.
Ling, W., N. Zhaohui, H. Ben, G. Leyi, L. Jianping, D. Huili, & Q. Jiaqi. (2008). Urinary IL-18
and NGAL as early predictive biomarkers in contrast-induced nephropathy after
coronary angiography. Nephron Clin Pract 108 (3):pp. c176-181, ISSN 1660-2110.
Louis, B. M., B. S. Hoch, C. Hernandez, N. Namboodiri, G. Neiderman, A. Nissenbaum, F. P.
Foti, A. Magno, G. Banayat, F. Fata, N. L. Manohar, & H. I. Lipner. (1996).
Protection from the nephrotoxicity of contrast dye. Renal Failure 18 (4):pp. 639-646,
ISSN 0886-022X
Meier, P., D. T. Ko, A. Tamura, U. Tamhane, & H. S. Gurm. (2009). Sodium bicarbonate-
based hydration prevents contrast-induced nephropathy: a meta-analysis. BMC
Med 7:pp. 23, ISSN 1741-7015.
252 Advances in the Diagnosis of Coronary Atherosclerosis

Merten, G. J., W. P. Burgess, L. V. Gray, J. H. Holleman, T. S. Roush, G. J. Kowalchuk, R. M.


Bersin, A. Van Moore, C. A. Simonton, 3rd, R. A. Rittase, H. J. Norton, & T. P.
Kennedy. (2004). Prevention of contrast-induced nephropathy with sodium
bicarbonate: a randomized controlled trial. JAMA 291 (19):pp. 2328-2334, ISSN
1538-3598.
Nikolsky, E., R. Mehran, D. Turcot, E. D. Aymong, G. S. Mintz, Z. Lasic, A. J. Lansky, E.
Tsounias, J. W. Moses, G. W. Stone, M. B. Leon, & G. D. Dangas. (2004). Impact of
chronic kidney disease on prognosis of patients with diabetes mellitus treated with
percutaneous coronary intervention. American Journal of Cardiology 94 (3):pp. 300-
305, ISSN 0002-9149.
Obert, D. M., P. Hua, M. E. Pilkerton, W. Feng, & E. A. Jaimes. (2011). Environmental
tobacco smoke furthers progression of diabetic nephropathy. American Journal of the
Medical Sciences 341 (2):pp. 126-130, ISSN 1538-2990.
O'Keefe, J. H., M. Abuannadi, C. J. Lavie, & D. S. Bell. (2011). Strategies for optimizing
glycemic control and cardiovascular prognosis in patients with type 2 diabetes
mellitus. Mayo Clinic Proceedings 86 (2):pp. 128-138, ISSN 1942-5546.
Parfrey, P. S., S. M. Griffiths, B. J. Barrett, M. D. Paul, M. Genge, J. Withers, N. Farid, & P. J.
McManamon. (1989). Contrast material-induced renal failure in patients with
diabetes mellitus, renal insufficiency, or both. A prospective controlled study. New
England Journal of Medicine 320 (3):pp. 143-149, ISSN 0028-4793.
Perkins, B. A., R. G. Nelson, B. E. Ostrander, K. L. Blouch, A. S. Krolewski, B. D. Myers, & J.
H. Warram. (2005). Detection of renal function decline in patients with diabetes and
normal or elevated GFR by serial measurements of serum cystatin C concentration:
results of a 4-year follow-up study. Journal of the American Society of Nephrology 16
(5):pp. 1404-1412, ISSN 1046-6673.
Pflueger, A., T. S. Larson, K. A. Nath, B. F. King, J. M. Gross, & F. G. Knox. (2000). Role of
adenosine in contrast media-induced acute renal failure in diabetes mellitus. Mayo
Clinic Proceedings 75 (12):pp. 1275-1283, ISSN 0025-6196.
Phisitkul, K., K. Hegazy, T. Chuahirun, C. Hudson, J. Simoni, H. Rajab, & D. E. Wesson.
(2008). Continued smoking exacerbates but cessation ameliorates progression of
early type 2 diabetic nephropathy. American Journal of the Medical Sciences 335
(4):pp. 284-291, ISSN 0002-9629.
Rosenstock, J. L., R. Bruno, J. K. Kim, L. Lubarsky, R. Schaller, G. Panagopoulos, M. V.
DeVita, & M. F. Michelis. (2008). The effect of withdrawal of ACE inhibitors or
angiotensin receptor blockers prior to coronary angiography on the incidence of
contrast-induced nephropathy. International Urology and Nephrology 40 (3):pp. 749-
755, ISSN 0301-1623.
Russo, D., A. Testa, L. Della Volpe, & G. Sansone. (1990). Randomised prospective study on
renal effects of two different contrast media in humans: protective role of a calcium
channel blocker. Nephron 55 (3):pp. 254-257, ISSN 0028-2766.
Saleem, S., F. G. Lakkis, & M. Martinez-Maldonado. (1996). Atheroembolic renal disease.
Seminars in Nephrology 16 (4):pp. 309-318, ISSN 0270-9295.
Shammas, N. W., M. J. Kapalis, M. Harris, D. McKinney, & E. P. Coyne. (2001).
Aminophylline does not protect against radiocontrast nephropathy in patients
Contrast-Induced Nephropathy in Patients with Type 2 Diabetes Mellitus
and Coronary Artery Disease: Update and Practical Clinical Applications 253

undergoing percutaneous angiographic procedures. Journal of Invasive Cardiology 13


(11):pp. 738-740, ISSN 1042-3931.
Shannon, M. (1999). Life-threatening events after theophylline overdose: a 10-year
prospective analysis. Archives of Internal Medicine 159 (9):pp. 989-994, ISSN 0003-
9926.
Shlipak, M. G., L. F. Fried, M. Cushman, T. A. Manolio, D. Peterson, C. Stehman-Breen, A.
Bleyer, A. Newman, D. Siscovick, & B. Psaty. (2005). Cardiovascular mortality risk
in chronic kidney disease: comparison of traditional and novel risk factors. JAMA
293 (14):pp. 1737-1745, ISSN 1538-3598.
Solomon, R. (2005). The role of osmolality in the incidence of contrast-induced nephropathy:
a systematic review of angiographic contrast media in high risk patients. Kidney
International 68 (5):pp. 2256-2263, ISSN 0085-2538.
Solomon, R. (2009). Preventing contrast-induced nephropathy: problems, challenges and
future directions. BMC Med 7:pp. 24, ISSN 1741-7015.
Solomon, R. J., M. K. Natarajan, S. Doucet, S. K. Sharma, C. S. Staniloae, R. E. Katholi, J. L.
Gelormini, M. Labinaz, & A. E. Moreyra. (2007). Cardiac Angiography in Renally
Impaired Patients (CARE) study: a randomized double-blind trial of contrast-
induced nephropathy in patients with chronic kidney disease. Circulation 115
(25):pp. 3189-3196, ISSN 1524-4539.
Solomon, R. J., R. Mehran, M. K. Natarajan, S. Doucet, R. E. Katholi, C. S. Staniloae, S. K.
Sharma, M. Labinaz, J. L. Gelormini, & B. J. Barrett. (2009). Contrast-induced
nephropathy and long-term adverse events: cause and effect? Clin J Am Soc Nephrol
4 (7):pp. 1162-1169, ISSN 1555-905X.
Spargias, K., E. Alexopoulos, S. Kyrzopoulos, P. Iokovis, D. C. Greenwood, A. Manginas, V.
Voudris, G. Pavlides, C. E. Buller, D. Kremastinos, & D. V. Cokkinos. (2004).
Ascorbic acid prevents contrast-mediated nephropathy in patients with renal
dysfunction undergoing coronary angiography or intervention. Circulation 110
(18):pp. 2837-2842, ISSN 1524-4539.
Staniloae, C. S., S. Doucet, S. K. Sharma, R. E. Katholi, K. R. Mody, J. T. Coppola, & R.
Solomon. (2009). N-Acetylcysteine added to volume expansion with sodium
bicarbonate does not further prevent contrast-induced nephropathy: results from
the cardiac angiography in renally impaired patients study. J Interv Cardiol 22
(3):pp. 261-265, ISSN 1540-8183.
Toprak, O., M. Cirit, M. Yesil, D. W. Byrne, N. Postaci, S. Bayata, K. M. Majchrzak, & E. Esi.
(2006). Metabolic syndrome as a risk factor for contrast-induced nephropathy in
non-diabetic elderly patients with renal impairment. Kidney and Blood Pressure
Research 29 (1):pp. 2-9, ISSN 1420-4096.
Vassallo, R., & J. J. Lipsky. (1998). Theophylline: recent advances in the understanding of its
mode of action and uses in clinical practice. Mayo Clinic Proceedings 73 (4):pp. 346-
354, ISSN 0025-6196.
Yoon, M. H., S. J. Tahk, H. M. Yang, J. S. Park, M. Zheng, H. S. Lim, B. J. Choi, S. Y. Choi, U.
J. Choi, J. W. Hwang, S. J. Kang, G. S. Hwang, & J. H. Shin. (2009). Comparison of
the intracoronary continuous infusion method using a microcatheter and the
intravenous continuous adenosine infusion method for inducing maximal
254 Advances in the Diagnosis of Coronary Atherosclerosis

hyperemia for fractional flow reserve measurement. American Heart Journal 157
(6):pp. 1050-1056, ISSN 1097-6744.
Zoungas, S., A. Patel, J. Chalmers, B. E. de Galan, Q. Li, L. Billot, M. Woodward, T.
Ninomiya, B. Neal, S. MacMahon, D. E. Grobbee, A. P. Kengne, M. Marre, & S.
Heller. (2010). Severe hypoglycemia and risks of vascular events and death. New
England Journal of Medicine 363 (15):pp. 1410-1418, ISSN 1533-4406.
14

Quantitative Coronary Angiography


in the Interventional Cardiology
Salvatore Davide Tomasello1, Luca Costanzo2
and Alfredo Ruggero Galassi1
1Department of Internal Medicine and Systemic Disease,

Catheterization Laboratory and Cardiovascular Interventional


Unit, Cannizzaro Hospital, University of Catania
2Department of Internal Medicine and Systemic Disease,

Clinical Division of Cardiology, Ferrarotto


Hospital, University of Catania
Italy

1. Introduction
Coronary angiography through selective injection of radiopaque contrast agent into the
coronary arteries is currently the gold standard for evaluating coronary artery disease
(CAD). Subjective visual estimation of lesion severity has been shown to be inadequate due
to high degrees of intraobserver and interobserver variability (Topol & Nissen, 1995). Since
the late 1980s methods and algorithms for quantitative coronary angiography (QCA) have
been developed in order to objectively quantify the extent of CAD (Serruys et al., 1984).
Furthermore, validation of specific QCA measurements associated with clinical outcomes
has led to their incorporation into various scoring systems and trials endpoint.

2. Two-dimensional QCA
This technique is based on contrast coronary angiograms obtaining parameters that quantify
objectively the coronary lumen measuring the significance of a coronary stenosis. Notably, a
single coronary angiography image gives only a two-dimensional (2D) radiographic image
while coronary arteries can be highly tortuous three-dimensional structures thus at least two
projections orthogonal to each other and both perpendicular to the analyzed coronary
segment are necessary to obtain the most complete data to avoid foreshortening or
underestimation of stenosis severity.
Most of the systems available nowadays are based on vessel edge detection algorithms to
designate the arterial lumen on conventional coronary angiograms providing, therefore, the
assessment of the lesion in an operator-independent way exploiting specific and dedicated
software. The earliest form of quantitative angiography was the application of calipers to
visually estimate percent diameter stenosis. In 1971 Gensini and colleagues described an
electronic caliper system in which the arterial borders of the lesion and normal segments are
manually defined by moving cursors (Gensini et al., 1971). In the years several generations
256 Advances in the Diagnosis of Coronary Atherosclerosis

of QCA systems have been developed based on edge detection algorithms to improve
measurement accuracy. First-generation algorithms have been introduced at the beginning
of the Eighties (Mancini et al., 1987; Reiber et al., 1985) improving the reliability of coronary
measurements (Spears et al., 1983); the main pitfall of these first generation systems was the
overestimation of the diameters of small vessels (Herrington et al., 1993a), an issue which
was improved in the second generation systems (diameter approximation \1.2 mm) and
producing more reliable results at the low end of the spectrum of vessel size (Hausleiter et
al., 1997). In more recent years cardiovascular X-ray imaging systems are equipped with
flat-panel (FP) detectors (Spahn et al., 2003) replacing the combination of image intensifier
and charge-couple device camera providing better image quality and enabling further image
enhancement. Novel QCA third-generation system which took advantage of digital flat-
panel (FP) detectors instead of the conventional image intensifier systems were better able to
determine smaller diameter vessels (Van Herck et al., 2004) and analyze complex lesion
morphology with irregular borders (Van der Zwet & Reiber, 1994). However differences in
results using FP systems compared with classic image intensifier-based imaging systems
were shown to be clinically not significant (Tuinenburg et al., 2006). In the common clinical
practice there are different computer systems for QCA that have been validated: among
them the most commonly employed are CAAS (PIE Medical, Maastricht, The Netherlands)
and QAngio XA (Medis, Leiden, The Netherlands) (Garrone et al., 2009).

3. Three-dimensional QCA
Currently, 2-dimensional (2D) QCA is the most commonly used and validated form of QCA.
However, 2D angiography technology has limitations because of vessel overlap and vessel
foreshortening during image acquisition. This is especially problematic in more complex
lesions such as bifurcations. 3D angiography utilizes standard 2D angiographic images
obtained at least 30 degrees apart and with minimal vessel overlap, and reconstructs 3D
images. Several 3D QCA programs are now available: among them the most commonly
employed are CardiOp-B (Paieon Medical Ltd. Park Afek, Israel) and CAAS 5 (Pie medical
Imaging, Maastricht, The Netherlands, and Medis, Leiden, The Netherlands) (Ng & Lansky,
2011). In these systems, the user defines the area of interest by defining the proximal end,
distal end and area of stenosis. However, the systems differ in their calibration methods.
CardiOp-B requires the operator to input the size of the catheter in the image into the
program, whereas the Medis and CAAS 5 systems automatically calibrates the images using
DICOM (Digital Imaging and Communications in Medicine) information embedded in the
angiographic images (Gradaus et al., 2006; Ramcharitar et al., 2008a). Once the image is
created, operators can manipulate the images by zooming in or rotating the image. This
allows the operator to have a better understanding of coronary anatomy and could also help
the operator find an optimal angle to image of a vessel for an intervention (Dvir et al., 2005;
Agostoni et al., 2008). An example of coronary after reconstructed with CardiOp-B system is
shown in figure 1.
3D QCA programs have been validated (Schuurbiers et al., 2009; Tsuchida et al., 2007) and
are now available to provide information of vessel size, percent diameter stenosis, minimal
lumen diameter, bifurcation angle and other QCA values. It was thought that 3D QCA
would have improved accuracy by resolving problems with vessel foreshortening and out of
plane magnification. However, comparisons of 3D QCA programs to conventional 2D QCA
programs have shown mixed results regarding the accuracy of this technology (Ramcharitar
Quantitative Coronary Angiography in the Interventional Cardiology 257

et al., 2008a; Dvir et al., 2005; Tsuchida et al., 2007; Meerkin et al., 2010; Wellnhofer et al.,
1999; Tu et al., 2010). One small study has compared the two current 3D QCA programs
using phantom models demonstrated that the CAAS 5 system may be more accurate than
the CardiOp-B system (Ramcharitar et al., 2008a).

Fig. 1. Reconstruction with 3-D CardiOp-B system of proximal left anterior descending
artery.

4. Materials and procedural details


The first step in order to perform a QCA analysis is to acquire high-quality angiographic
images of the coronary artery segment of choice using a standard and reliable technique.
Coronary angiography actually provides a two-dimensional luminogram of a three-
dimensional structure, formed not only of the lumen itself but also of the vessel wall.
Therefore, coronary angiography has important limitations that can have a significant
impact on the results of QCA (Topol & Nissen, 1995). It is thus clear that coronary
angiography cannot analyze coronary remodeling mechanisms, that is, the outward or
inward changes in total vessel area that may not correspond to outward or inward changes
in lumen area typical of atherosclerosis in an effective way (Glagov et al., 1987).
Furthermore, to get coronary angiographic images that profile the exact coronary anatomy
with as much details as possible and avoid foreshortening or underestimation of stenosis
severity, it is necessary also that the C-arm tube is directly orthogonal to the coronary
segment analysed, and it is advisable to get at least two projections orthogonal to each other
and both perpendicular to the coronary segment in order to obtain the most complete data.
Selection of a coronary image for analysis depends on several factors linked to the limits of
radiographic coronary images. The real issue is the choice of the sequence where the
stenosis has the minimal foreshortening and the minimal overlap with other structures (e.g.,
other coronary arteries, bifurcation branches, catheters, or radiopaque surrounding
structures such as a calcified mitral valve). The greater is the contrast between the
radiopaque contrast-filled coronary artery and the radiotransparent background, the greater
258 Advances in the Diagnosis of Coronary Atherosclerosis

are the accuracy of QCA analysis and reliability of the algorithm for the automated
detection and reconstruction of the lumen edges. That is why it is also important that the
coronary images are obtained when the patient is in deep inspiration, which increases the
radiotransparent lung fields surrounding the heart and thus the possibility that the stenosis
is in front of this radiotransparent field. It is also important that the distal tip of the coronary
angiography catheter is present in the sequence, at least in the initial frames.
Indeed, the tip of the catheter, whose diameter in French or millimeters is known, is the
basic tool for calibration (i.e., to estimate the proportion between pixels in the digitalized
images and mm of length). Finally, intracoronary nitroglycerine (e.g., 200 μg) might be
administered to obtain coronary vasodilatation and avoid to mistakenly confounding
vasospasm with atherosclerotic processes.
After choosing the appropriate run, a single static image (i.e., frame) is selected which
should be chosen in end-diastole, when coronaries tend to “stretch” along their course on
the released heart muscle without being subject to contraction or squeezing, even in the case
of intra-myocardial course. Regardless of the type of software, the analysis proceeds
similarly and the following steps consist in: digitization, image calibration and arterial
automatic contour detection. Advances have been made to each of these steps over the last
several decades. The process of digitization from cine film using cine video converter is
obsolete and it has been replaced by digital systems with direct digital input to a video
camera. The analogic video output signal is modified electronically and results in a greater
contrast difference in the parts with high X-ray absorption and lower contrast differences in
areas with low X-ray absorption, significantly improving image quality. Image calibration is
often done with contrast-filled injection catheters (Figure 2), and can be problematic due to
variable image quality and out-of-plane magnification that can be overcome by using
biplane calibration.

Fig. 2. Image calibration using contrast-filled injection catheter.


Quantitative Coronary Angiography in the Interventional Cardiology 259

A central line is usually then drawn by hand along the stretch of the catheter tip and the
software automatically recognizes its margins by using specific algorithms that, by means of
digital images, recognize the change from radiopaque pixels (black or dark grey) to radio-
transparent pixels (light grey or white) according to a densitometric analysis. The software
then transforms every pixel into a square with sides characterized by known dimensions
(mm). This parameter is then employed to measure the coronary segment of interest that
must be focused on the stenosis or lesion undergoing treatment. The proximal and distal
coronary segments should be relatively free of disease and are referred to as reference
segments. Afterwards, a central line is traced manually and the software automatically
recognizes the margins of the coronary segment under consideration taking advantage of
the automatic contour detection .
This procedure determines the vessel edge based on the weighted sum of the first and
second derivative functions of the brightness values of scanlines perpendicular to the
centerline, which in turn undergoes an analysis that determines the optimal contour path
along the segment (Figure 3).

Fig. 3. After having traced a central line the software recognizes the margins elaborating
automatically the contours.
A further step allows, always automatically, the reconstruction of the hypothetically normal
coronary lumen. An algorithm then creates a line of the coronary margin that interpolates
the coronary segment considered free from illness located proximally and distally to the
region of interest with the ones of the region of interest. The algorithm then reconstructs the
reference coronary segments (i.e., those apparently free from disease) (Figure 4).
This technique is essentially based on the calculation of a mean value of the diameters of the
lumen in the segments of reference located upstream and downstream to the lesion. It
appears thus clear the importance of including two coronary segments in the QCA analysis,
one proximal and one distal to the target segment and both angiographically-free from
significant disease. These steps produce different parameters, some measured directly and
others derived with interpolation techniques.
260 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 4. Software algorithm reconstructs the reference coronary segment.

5. Parameters
Coronary lesions can be assessed qualitatively and quantitatively, in terms of severity of the
lesion itself. The qualitative evaluation is based on the visual estimation of the lesion and it
depends very much on the operator’s experience. The quantitative evaluation, based on
QCA, allows instead to obtain numeric parameters that are much more independent from
the operator acquiring the images or the one performing the analyses. The coronary
parameters of major interest are summarized in Table 1.

Range
Parameter commonly Meaning
used
Minimal Luminal Diameter The smallest lumen diameter in the segment
0–6.00 mm
(MLD) of interest
Reference Vessel Diameter The averaged diameter of the coronary
1.5–6.0 mm
(RVD) assumed without atherosclerotic disease
Length of the stenosis as measured by 2 points
where the coronary margins change direction,
Lesion length 0–60.0 mm creating a shoulder between the
angiographically normal subsegment and the
diseased subsegment
Acute gain 0–4.0 mm Post-procedural MLD – pre-procedural MLD
Late loss (LL) 0.10 to 3.00 Post-procedural MLD – MLD at follow-up
Diameter stenosis (DS) 0–100% (RVD-MLD)/RVD
DS >50% at follow-up coronary angiography
Binary restenosis (BR) Yes or no
in the treated coronary segment
Table 1. Main parameters obtained with QCA (adapted from Garrone et al., 2009)
Quantitative Coronary Angiography in the Interventional Cardiology 261

Additional tool of QCA are also available, indeed it is possible analysis of the stent edges
after stent implantation. In fact, the radiopaque stent can generally be visualized during
angiographic sequence when the contrast medium has not yet been injected in the coronary
vessel. By comparing this image with the frame selected for the QCA analysis, it is possible
to determine the two edges, proximal and distal, of the implanted stent. The software then
enables to mark the edges with a line transversal to the vessel lumen in order to
automatically calculate the segment where the stent has been implanted and to calculate the
5-mm segments that lie proximal and distal to the stent. Therefore, it is possible to
determine MLD and DS for each one of the three segments considered.
Several angiographic phenomena can bias QCA and lead to overestimation or
underestimation of QCA parameters such as acute or chronic thrombus, extensive calcium
deposits, dissections, diffuse disease, and slow flow, therefore it is of paramount importance
to add a qualitative analysis for lesion features to any QCA analysis (Garrone et al., 2009).

6. Applications
QCA can be employed in clinical practice during diagnostic coronary angiography (online),
to have an objective and independent parameter for the assessment of stenosis severity as
the human eye does not have the resolution capability of software. Usually, visual
interpretation of the severity of a coronary stenosis is expressed in intervals of percentage of
stenosis while QCAproduces a single specific measure for DS, improving the accuracy and
reproducibility of the severity assessment (Kalbfleisch et al., 1990). It has been shown that
the visual assessment tends to underestimate stenoses<50% and to overestimate those >50%
(Fleming et al., 1991) therefore the online use of QCA assessing the lesion length and RVD
might allow to obtain objective parameters to decide which specific measures (mainly
diameter and length) of the device (e.g., balloon or stent) should be chosen.
In the field of clinical research QCA allows serial analysis of the same coronary segment
over time in an indipendent way. In trials on coronary devices typically at least three QCA
runs are performed, one before treatment, another immediately after treatment, and a final
one at angiographic follow-up, after a respecified time.
In recent years late luminal loss and binary restenosis have taken an important role in
several studies aimed at evaluating PCI devices, in particular, stents. These values, more
specific and more prevalent of the clinically relevant end-point of target lesion
revascularization (TLR), allow researchers to compare different types of devices in clinical
trials, with greater statistical power, and thus with fewer patients. In fact, late luminal loss is
a continuous parameter and is in itself statistically more powerful and precise, while binary
restenosis, despite being a value present or absent as the TLR, occurs with an incidence
roughly two times higher than the incidence of TLR. Therefore QCA can reduce the number
of patients needed in a clinical trial to obtain significant differences between different
devices.
Another interesting application is the study of plaque progression/regression that is based
on at least two QCA runs made over a pre-specified interval of time, usually at least 1 year.
The change in MLD of the coronary segments of interest is quantified after a given
pharmacologic regimen that purports to act on the development or progression
ofatherosclerotic disease.
262 Advances in the Diagnosis of Coronary Atherosclerosis

7. Prediction of functional significance of a stenosis


2D-QCA systems have substantially improved reproducibility and accuracy of lesion
dimension assessment, especially percent diameter stenosis, however its use for the
prediction of functional significance of the lesion was shown to have only a modest
correlation with the more accurate Flow Fractional Reserve (FFR) (Bartunek et al., 1995;
Fischer et al., 2002). Conversely, 3D-QCA has shown reasonable accuracy in predicting FFR
< 0.75 in patients with visually determined intermediate stenosis (Saad et al., 2009).
Recently, Yong and co-workers investigated the accuracy of 3D-and 2D-QCA measurements
in predicting both FFR <0.75 and ≤0.80, and compared the correlation of FFR with 3D- and
2D-QCA measurements in 63 coronary lesions (Yong et al., 2011). Interesting, the
relationship between FFR and apparent stenosis severity was found to be curvilinear. Of all
measurements of lesion severity obtained by 3D-QCA, MLA best correlated with FFR (R =
0.63, P < 0.001), and was the most accurate predictor of FFR < 0.75 (C statistic 0.86, P =
0.001). Of 2D-QCA measurements, MLD correlated best with FFR (R = 0.58, P < 0.001), and
best predicted FFR <0.75 (C statistic 0.80, P < 0.001). Overall, 3D-QCA showed a non-
significant trend towards more accurate prediction of FFR than 2D-QCA, especially in
intermediate lesions. The authors then concluded that both 3D- and 2D-QCA were less
accurate in intermediate lesions and in predicting FFR ≤0.80 than in predicting FFR < 0.75
(Yong et al., 2011).

8. Special coronary lesion subset: chronic total occlusion and bifurcations


8.1 QCA in chronic total occlusion
Chronic total occlusions (CTOs) of the coronary arteries are one of the most technically
challenging lesion subsets for interventional cardiologists due to the most significant
difficulty in crossing the lesion with guidewire compared with other lesions subset (Di
Mario et al., 2007). This might be considered a failure of conventional 2D-angiography to
properly represent stump morphology and true lesion geometry (Safian et al., 1988;
Kinoshita et al., 1995). Moreover CTOs are often long lesions length, with severe
calcifications and tortuosity (Di Mario et al., 2007), therefore the evaluation of these
complex geometric factors is difficult with conventional 2D angiography, often leading
to misjudgement of the potential and proper method of treatment (Herrington et al,
1993b).
Recently, Dvir and colleagues evaluated real-time 3D reconstruction of CTOs with CardiOp-
B system in a series of 58 patients (61 CTOs) (Dvir et al., 2008). Authors reported a 3D
reconstruction success rate of 83% with improved visualization of stump area and/or
missing segment. Importantly, in 92% of the successful 3D reconstructions, the artery path
in the lesion area could be delineated and in 95% of cases, in which post-stenting 3D
reconstruction performed, the vessel path was similar to the lesion path suggested before
stenting. Interestingly, the mean stenosis area was significantly smaller in the 3D
reconstructions vs. 2D images (94 6 5.1% vs. 99 6 0.3%, P < 0.001) and the mean lesion length
was significantly shorter (15.3 6 7.4 mm vs. 20.9 6 8 mm, P < 0.001). A possible explanation
proposed by the Authors was that in these highly complex lesions, where lesion path was
not visible, the 2D measurements were severely inaccurate. Thus 3D reconstructions may be
considered as a useful tool in CTO procedures to improve assessment of CTO lesions
characteristics (Dvir et al., 2008).
Quantitative Coronary Angiography in the Interventional Cardiology 263

8.2 Bifurcations
With the expanding practice of stenting coronary bifurcation lesions worldwide , the need
for reliable, standardized and reproducible quantitative bifurcation analyses became
apparent. A fundamental challenge in assessing bifurcations using quantitative methods is
in acquiring the entire bifurcation lesion without significant foreshortening or vessel and
side branch overlap. While current QCA standards require a minimum of two orthogonal
views more than 30° apart, optimal visualization of the bifurcation lesion (parent vessel and
side branch) is typically available in one best single view. When QCA is applied using a
standard operating procedure, i.e. by including in the analyzed segment the proximal main
vessel (PMV) and distal main vessel (DMV), the RVD function takes an average of the
‘‘normal’’ segments of the proximal and distal vessel. Tapering, however, causes the
proximal RD and thus the percentage diameter reduction of lesions proximal to the
bifurcation to be underestimated, while the distal RD and %DS of lesions distal to the
bifurcation will be overestimated. If on-line QCA is used for guidance during a procedure,
this can lead to inappropriate balloon selection, with significant under sizing of balloons
used for dilatation of the proximal segment and equally, over sizing of balloons used for the
distal segment of the main vessel (MV). The operator must manually redraw the proximal
segment within the MV and is forced to tediously correct for the contours of the lesion, a
possible source of inaccuracy and subjectivity. Even with optimal angiographic views, the
main issues of conventional QCA in the analysis of bifurcation lesions are: defining the true
reference vessel size of both the parent vessel and its side branch; the assessment of side-
branch (SB) stenoses when truly ostial. In the non-diseased state, the RD of the main vessel
is reduced from proximal to distal relative to the side branch. It has been shown that the
change in RD of the main vessel across the side branch is a function of the size of the PMV,
the DMV and that of the SB. Murray was the first to describe the mother/daughter-vessel
relations in vascular bifurcations, in terms of a cost function: i.e., the sum of function power
loss and metabolic power dissipation proportional to blood flow. Murray described the
optimal conditions for vascular bifurcation, known as Murray’s law, which states that the
cube of the radius of the mother vessel equals the sum of the cubes of the radii of the
daughter vessels: Dm3=Dd13+Dd23 (Murray, 1926). Afterwards, a simplified equation relating
the size of the proximal and distal main vessel and the side branch, in the non-diseased
state, has been described by a constant [D1= 0.687 (D2 + D3 + …)] (Kassab, 2006; Kamiya &
Takahashi, 2007) such that the reference size of any one segment can always be derived if
the two other segment sizes are known (Finet et al., 2007). Standard QCA algorithms are
designed to detect vessel contours assuming minimal vessel tapering; thus, the reference
vessel dimensions are inherently inaccurate when applied to bifurcation lesions. Several
techniques have been used to attempt to overcome this limitation but remain problematic
(Lansky et al., 2009).

8.2.1 Dedicated bifurcation QCA algorithms


Currently two dedicated bifurcation QCA software packages are commercially available
(Medis medical imaging systems, Leiden and The Netherlands and Pie Medical, Maastricht,
The Netherlands). The main issue for the application of QCA in bifurcating segments
include:
- minimal user interaction in the selection and processing of the coronary segments to be
analysed;
264 Advances in the Diagnosis of Coronary Atherosclerosis

- minimal editing of the automatically determined results;


- short analysis time;
- providing highly accurate and precise results, with small systematic and random errors
- providing suitable reporting and data exchange mechanisms.
The Medis medical imaging systems. This system contains two bifurcation models: a T-
shape bifurcation model (suitable for bifurcations with a standard side branch structure)
(Figure 5 A) and a Y-shape bifurcation model (suitable for bifurcations with distal branches
of equal size) (Figure 5 B).

(Adapted from Tuinenburg et al., 2011).

Fig. 5. T-shape model (A) and Y-shape model (B) showing the segments, proximal delimiter,
and sections terminology. For each model, the segment generated four segments that
represent the building blocks of the models. A) Using the T-shape model, the arterial and
reference diameters of the ostium of the side branch and the whole main section (including
the transition within the bifurcation core) can be accurately determined. B) Using the Y-
shape model, the arterial and reference diameters up to the carinal point and in the distal 1
and 2 sections can be determined accurately.
Quantitative Coronary Angiography in the Interventional Cardiology 265

The particular advantage of these models is that they combine the proximal and two distal
vessel segments with the bifurcation core, resulting in a total of two or three sections, all
derived from one analysis procedure, such that each of these sections has its own diameter
function and associated parameter data.
Step 1. Contour detection. Three path-line points have to be selected to define the arterial
bifurcation segment: a start point in the proximal parent vessel (PV), and end points
in the distal PV and the SB. From these points, two pathlines are constructed
followed by the automated detection of the arterial contours of all three vessel
segments using an algorithm.
Step 2. Lesions analysis. The bifurcation core is defined automatically as the area between
the first diameter of the distal PV segment, and the interpolated contour between
the proximal and distal PV segments.
In the T Bifurcation model, the proximal and distal PV and core segments are combined to
define the first section and the SB segment forms the second section. For the entire PV
section the arterial diameter function is calculated following the conventional Medis
straight analysis approach, while for the SB section the Medis ostial analysis approach is
followed.
For the Y bifurcation model, the core segment is defined as the area between the
automatically determined proximal boundaries of the proximal segment and the carinal
point. Excluding the core segment, three sections are defined with each arterial diameter
functions calculated using the conventional straight analysis approach.
Step 3. Result reporting. The results of the bifurcation analysis are reported individually for
the proximal segment (combining the proximal PV and core segments), the distal
PV segment, the SB segment, and the ‘‘combined lesion’’ results (combined
proximal, core, and distal PV). Conventional angiographic parameters will be
reported including the obstruction, reference, minimum, maximum, and mean
diameters and areas, the percent diameter and area stenosis, as well as the vessel
and lesion lengths.
The Pie Medical Analysis Principles: CAAS 5 QCA bifurcation methodology. In this
system the bifurcated vessel is considered to be a single entity, including the central
bifurcation area divided in 10 segments when stented (Ramcharitar S, 2008) (Figure 6).
In this analysis, the segments 1, 4 and 6 are 5 mm beyond the stent edge. The main
vessel stent is divided at the carina as that proximal and distal corresponding to
segments 2 and 3 respectively, with the side branch stent confined to segment 5. The
central portion, segment 7 is an independent region called the Polygon of Confluence
(POC) that is unique to the bifurcation and represents a new quantification modality.
Segment 8 is an ostial region of side branch including 5 mm distal from the end of
bifurcation. The CAAS 5 quantitative analysis of bifurcations can be summarized in six
steps:
1. Contour detection, that is performed in a semi-automated process employing either
manually drawn initial pathline, or three user-defined points (one at the proximal side
of the main vessel and two at both distal ends of the bifurcated branches). The
algorithm automatically calculates path lines and the contours. The software assumes
the bifurcated vessel is a single object delineated by a left, middle and right contour,
specifically making no further assumptions.
266 Advances in the Diagnosis of Coronary Atherosclerosis

1 – Proximal Edge (5 mm) 6 – Distal Edge Side (5 mm)


2 – Proximal Main Stent 7 – Polygon of Confluence
3 – Distal Main Stent 8 – Ostium of the Side Branch (5 mm)
4 – Distal Edge Main (5 mm) 9 – MV Stent + Edges
5 – Side Branch Stent 10 – SB Stent + Distal Edge
Fig. 6. The 10 segments used in CAAS (Adapted from Ramcharitar et al., 2008b)
2. Construction of the centreline and defining the Point of Bifurcation (POB). The Point of
Bifurcation (POB) is defined as the point where all the centrelines meet; the centrelines
being the lines through the middle of the vessel. The POB is defined as the midpoint of
the largest circle that can be fitted in the bifurcation area, touching all three contours.
The intersections between this circle and the centrelines identify the start and end
positions of the bifurcation region the boundaries of a new measurement entity the
POC (Figure 7).
3. Defining the POC. The POC represents the smallest possible independent region that
behaves differently from a single vessel segment. It is defined on the 2D radiographic
image as the area or region that encompasses the start and the end of the bifurcation
region. Regions outside of the POC behave similar to single vessels in classic QCA. The
polygon shape is created by connecting begin and end boundaries of the POC by the
luminal borders of the vessel. Depending on the type of bifurcation it can have different
shapes, such as a pentagon. The plaque area can be evaluated against the actual
luminal area or volume to give the percentage area or volume stenosis as a single value
for the POC after reconstruction the vessel to its healthy situation. Furthermore the
absolute POC area can be compared between pre-, post- and follow-up in clinical
studies as a measure for procedural success (Figure 7).
Quantitative Coronary Angiography in the Interventional Cardiology 267

Fig. 7. Polygon of confluence and point of bifurcation in CAAS system (Adapted from
Ramcharitar, 2008b)
4. Determination diameter and reference values within the bifurcation. Traditional diameter
measurements can also be obtained. However, unlike the branches where the diameter
measurements are determined by the distance between left and right border, it is not
straightforward inside the POC. In order to derive such data within the POC the
“Minimum Freedom” approach must be employed. In this approach shortest distances
between a centreline point and the boundaries are determined, and the distance
between these points used as best estimate for the diameter within the POC for that
location along the centreline. The MLD at the bifurcation can be found using this
approach by walking through points along the termini of the bifurcation to the centre
point within the polygon of confluence. The reference diameter outside the POC is
determined from the edges of the ‘healthy’ part of the vessel. It is based only on the
characteristics of the branch itself as in the CAAS single vessel software.
5. Determining the bifurcation angulation. For the calculation of the angles between the
arterial branches, a method that employs lines derived from the circle used for the
definition of the “POC” can be used. The intersection positions of the circle with the
centrelines are used as the centres for new circles. These new circles have the same
radius as the circle within the “POC” and are not dependent on the mean vessel
diameter of the specific branch.
6. Report. Different versions of the bifurcation analysis software are available for clinical
and core lab use. Available analysis methods include automatic reference obstruction
analysis, local reference analysis, user defined subsegments as well as the 10
segmental analysis method to analyse the stented bifurcations, post-procedure and at
follow-up.

8.2.2 3D QCA in bifurcation


3D-QCA analysis is able to follow the complex arterial curvilinear structure of the
bifurcation and to select the most appropriate view to avoid foreshortening in the region of
interest. 3D-QCA, as compared to conventional two-dimensional QCA, allows a more
appropriate imaging of bifurcation geometry, with chance of an online assessment of the
bifurcation and deviation angle values, which may help the operator to choose the best
strategy to approach each lesion (Galassi et al., 2010). Although there are multiple software
268 Advances in the Diagnosis of Coronary Atherosclerosis

programs dedicated for 3-dimensional QCA, only two software packages are dedicated for
bifurcation analysis: The CardiOp-B system (Dvir et al., 2007; Galassi et al., 2010) and the
recent CAAS 5 QCA 3D bifurcation software (Onuma et al., 2011).
CAAS 3-D QCA, while following the already described algorithm based on vessel size
adjusted vectors, calculates BA values in 3-D space without overlap, therefore theoretically
more precisely than 2-D QCA. On the other hand, in CardiOp-B angle values were
computed between the weighted vectors of direction of the respective vessel segments. The
weighted vector is calculated as the sum of two vectors, each of them connecting the
bifurcation point with distal points along the vessel centre line, distant by 5 mm and 10 mm,
respectively. This algorithm was employed in a recent substudy of the SYNTAX trial, the
first ever to describe the 3-D angulation of the left main before and after intervention and its
impact on 1-year clinical outcome (Girasis et al., 2010a). When it comes to the quantification
of stenosis, CardiOp-B provides diameter-derived and cross-sectional (densitometric) data
together with the lesion length; lesion markers can be manipulated, in order to relocate the
region of interest across the main vessel or from the PMV into the SB. (Figure 8)

Fig. 8. An example of coronary bifurcation lesion reconstructed with 3D- CardiOp-B software.
Quantitative Coronary Angiography in the Interventional Cardiology 269

In CAAS 5, a 3-D model of the central bifurcation area is constructed taking into account the
fact that the contour information obtained from the 2-D projections may contain vessel
overlap, since the bifurcated vessel might be partly obscured in at least one of the
projections. To overcome this problem the 3-D cross-section shape is created using virtual
vessel contours by virtue of information derived outside the bifurcation region (Onuma,
2011).
Cross-sectional area values are calculated on the assumption that the vessel has an elliptical
cross section based on the luminal diameters from the two different 2-D projections; this
elliptical cross-section is the primary measured parameter. The equivalent luminal diameter,
minimum luminal diameter and maximal luminal diameter curves are calculated based on a
circularity assumption. Reference vessel lines, up to the entrance of the POC and within the
POC, are based on the 2-D approach adapted to 3-D.
So far, there is not direct comparison between these two QCA bifurcation software
packages, both of which recostruct from two projections, thus the issue of overlapping
vessels in tortuous bifurcation lesions may still be a problem. Particularly, there are two
special challenging situations in QCA analysis: a diffusely diseased bifurcation region
with no apparent healthy reference and an ostial stenosis in a short left main trunk
(Girasis, 2010 b). Based on the scaling laws of Murray (Murray, 1926) and Finet (Finet,
2007), the size of any bifurcation vessel segment could be determined, if the reference of
the other two vessel segments is known. This is not the case in a diffusely diseased
bifurcation region; however, extrapolation of the reference of the PMV even on the basis
of less accurate RVD values for the DMV and SB could avoid a gross underestimation of
the PMV true size (Girasis, 2010 b).
In the case of an ostial stenosis in a short left main stem, both contour detection and
sizing could be challenging. Manual contour corrections would not be unreasonable,
especially if there is overlap with the angiographic catheter. For the RVD determination,
either a user-defined reference outside the obstructions boundaries but within the left
main, or a value back-calculated from the distal branches reference values can be chosen
(Girasis, 2010 b).
Altough 3-D new techniques allow more extensive analyses for coronary bifurcations, there
are several limitations such as reproducibility, accuracy and standard reference values for
special lesion subsets that need further studies.

9. Conclusion
Although QCA was born more than 20 years ago and still burden well established
limitations, it remains a useful tool in clinical research, whereas its role in clinical practice
is mainly complementary to standard coronary angiography and other imaging
techniques, such as computed tomography (CT), intravascular ultrasound (IVUS), virtual
histology and optical coherence tomography. In recent years QCA has advanced to
provide more accurate measurements and descriptions of the coronary vascular tree and
still continues to evolve as 3D coronary angiographic techniques provide more detailed
descriptions of complex lesions such as bifurcations, eccentric lesions and chronic total
occlusions. However standard values in these lesions will need to be determined and
validated.
270 Advances in the Diagnosis of Coronary Atherosclerosis

10. References
Agostoni, P.; Biondi-Zoccai, G. & Van Langenhove, G. (2008). Comparison of assessment of
native coronary arteries by standard versus three-dimensional coronary
angiography. Am J Cardiol 102:272–279.
Bartunek, J.; Sys, S.U. & Heyndrickx, G.R.; (1995). Quantitative coronary angiography in
predicting functional significance of stenoses in an unselected patient cohort. J Am
Coll Cardiol 26:328–334.
Di Mario, C.; Werner, G.S. & Sianos, G. On behalf of the Euro CTO Club (2007). European
perpective in the recanalizzation of Chronic Total Occlusion (CTO): consensus
document from the Euro CTO Club. Eurointerv 3:30-43.
Dvir, D.; Marom, H. & Guetta, V. (2005). Three-dimensional coronary reconstruction from
routine single plane coronary angiograms: in vivo quantitative validation. Int J
Cardiovasc Intervent 7:141–145.
Dvir, D.; Marom, H. & Assali, A. (2007). Bifurcation lesions in the coronary arteries: early
experience with a novel 3-dimensional imaging and quantitative analysis before
and after stenting. EuroIntervention 3:95-9.
Dvir, D.; Assali, A. & Kornowski, R. (2008). Percutaneous coronary intervention for chronic
total occlusion: novel 3-dimensional imaging and quantitative analysis. Catheter
Cardiovasc Interv 71:784-789.
Fleming, R.M.; Kirkeeide, R.L. & Smalling, R.W. (1991). Patterns in visual interpretation of
coronary arteriograms as detected by quantitative coronary arteriography. J Am
Coll Cardiol 18:945–951.
Finet, G.G.M.; Perrenot, B. & Rioufol, G. (2007). Fractal geometry of arterial coronary
bifurcations. A quantitative coronary angiography and intravascular ultrasound
analysis. EuroInterv 3:490-498.
Fischer, J.J.; Samady, H. & McPherson, J.A. (2002). Comparison between visual assessment
and quantitative angiography versus fractional flow reserve for native coronary
narrowings of moderate severity. Am J Cardiol 90:210–215.
Galassi, A.R.; Tomasello, S.D. & Capodanno, D. (2010). A novel 3-d reconstruction system
for the assessment of bifurcation lesions treated by the mini-crush technique. J
Interv Cardiol 23:46-53.
Garrone, P.; Biondi-Zoccai, G. & Salvetti, I. (2009). Quantitative coronary angiography in the
current era: principles and applications. J Interv Cardiol 22:527-536.
Gensini, G.G.; Kelly, A.E. & DaCosta, B.C.B. (1971). Quantitative angiography: The
measurement of coronary vasomobility in the intact animal and man. Chest 60:522-
530.
Girasis, C.; Serruys, P.W. & Onuma, Y. (2010a). 3-Dimensional Bifurcation Angle Analysis in
Patients With Left Main Disease A Substudy of the SYNTAX Trial (SYNergy
Between Percutaneous Coronary Intervention With TAXus and Cardiac Surgery).
JACC Cardiovasc Interv 3:41-48.
Girasis, C.; van Jeuns, R.J. & Onuma, Y. (2010b). Essentials of quantitative angiography for
bifurcation lesions. Eurontervention Supplement Vol. 6 (Supplement J) J36-J43.
Glagov, S.; Weisenberg, E. & Zarins, C.K. (1987). Compensatory enlargement of human
atherosclerotic coronary arteries. N Engl J Med 316:1371–1375.
Quantitative Coronary Angiography in the Interventional Cardiology 271

Gradaus, R.; Mathies, K.; Breithardt G, Bocker D (2006). Clinical assessment of a new real
time 3D quantitative coronary angiography system: evaluation in stented vessel
segments. Catheter Cardiovasc Interv 68:44–49.
Hausleiter, J.; Jost, S. & Nolte, C.W. (1997). Comparative in vitro validation of eight first- and
second-generation quantitative coronary angiography systems. Coron Artery Dis
8:83–90.
Herrington, D.M.; Siebes, M. & Walford, G.D. (1993a). Sources of error in quantitative
coronary angiography. Cathet Cardiovasc Diagn 29:314–321.
Herrington, D.M.; Siebes, M. & Sokol, D.K. (1993b). Variability in measures of coronary
lumen dimensions using quantitative coronary angiography. J Am Coll Cardiol
22:1068–1074.
Kalbfleisch, S.J.; McGillem, M.J. & Pinto, I.M. (1990). Comparison of automated quantitative
coronary angiography with caliper measurements of percent diameter stenosis. Am
J Cardiol 65:1181–1184.
Kamiya, A. & Takahashi, T. (2007). Quantitative assessments of morphological and
functional properties of biological trees based on their fractal nature. J Appl Physiol
102:2315–2323.
Kassab, G.S. (2006). Scaling laws of vascular trees: Of form and function. Am J Physiol Heart
Circ Physiol 290:894–903.
Kinoshita, I.; Katoh, O. & Nariyama, J. (1995). Coronary angioplasty of chronic total
occlusions with bridging collateral vessels: Immediate and follow-up outcome
from a large single center experience. J Am Coll Cardiol 26:409–415.
Lansky, A.; Tuinenburg, J. & Costa, M. (2009). European Bifurcation Angiographic Sub-
Committee. Quantitative Angiographic Methods for Bifurcation Lesions: A
Consensus Statement from the European Bifurcation Group. Catheter Cardiovasc
Interv 73:258-266.
Mancini, G.B.; Simon, S.B.; & McGillem, M.J. (1987). Automated quantitative coronary
arteriography: morphologic and physiologic validation in vivo of a rapid digital
angiographic method. Circulation 75:452–460.
Meerkin, D.; Marom, H. & Cohen-Biton, O. (2010). Three-dimensional vessel analyses
provide more accurate length estimations than the gold standard QCA. J Interv
Cardiol 23:152–159.
Murray, C.D. (1926) The physiological principle of minimum work. I. The vascular system
and the cost of blood volume. Proc. Natl Acad. Sci. USA 12:207-214.
Ng, V.G. & Lansky, A.J (2011). Novel QCA methodologies and angiographic scores. Int J
Cardiovasc Imaging 27:157-165.
Onuma, Y.; Girasis, C. & Aben, J.P. (2011). A novel dedicated 3-dimensional quantitative
coronary analysis methodology for bifurcation lesions. Eurointervention [Ahead of
Print].
Ramcharitar, S.; Daeman, J. & Patterson, M. (2008a). First direct in vivo comparison of two
commercially available three-dimensional quantitative coronary angiography
systems. Catheter Cardiovasc Interv 71:44–50.
Ramcharitar, S.; Onuma, Y. & Aben, J.P. (2008b). A novel dedicated qualitative coronary
analysis methodology for bifurcation lesions. Eurointervention 3:553–557.
272 Advances in the Diagnosis of Coronary Atherosclerosis

Reiber, J.H.; Serruys, P.W. & Kooijman, C.J. (1985). Assessment of short-, medium-, and
long-term variations in arterial dimensions from computer-assisted quantitation of
coronary cineangiograms. Circulation 71:280–288.
Saad, M.; Toelg, R. & Khattab, A.A. (2009). Determination of haemodynamic significance of
intermediate coronary lesions using three-dimensional coronary reconstruction.
Eurointervention 5:573–579.
Safian, R.D.; McCabe C.H. & Sipperly, M.E. (1988). Initial success and long-term follow-up
of percutaneous transluminal coronary angioplasty in chronic total occlusions
versus conventional stenoses. Am J Cardiol 61:23G–28G.
Schuurbiers, J.C.; Lopez, N.G. & Ligthart, J. (2009). In vivo validation of CAAS QCA-3D
coronary reconstruction using fusion of angiography and intravascular ultrasound
(ANGUS). Catheter Cardiovasc Interv 73:620–626.
Spahn, M.; Heer, V. & Freytag, R. (2003). Flat-panel detectors in X-ray systems. Radiologe
43:340–350.
Serruys, P.W.; Reiber, J.H.C. & Wijns, W (1984). Assessment of percutaneous transluminal
coronary angioplasty by quantitative coronary angiography: Diameter versus
densitometric area measurements. Am J Cardiol 54:482–488.
Spears, J.R.; Sandor, T. & Als, A.V. (1983). Computerized image analysis for quantitative
measurement of vessel diameter from cineangiograms. Circulation 68:453–461.
Topol, E.J. & Nissen, S.E. (1995). Our preoccupation with coronary luminology. The
dissociation between clinical and angiographic findings in ischemic heart disease.
Circulation 92:2333–2342.
Tsuchida, K.; van der Giessen, W.J. & Patterson M (2007) In vivo validation of a novel three-
dimensional quantitative coronary angiography system (CardiOp-B): comparison
with a conventional two-dimensional system (CAAS II) and with special reference
to optical coherence tomography. EuroIntervention 3:100–108.
Tu, S.; Koning, G. & Jukema, W. (2010). Assessment of obstruction length and optimal
viewing angle from biplane X-ray angiograms. Int J Cardiovasc Imaging 26:5–17.
Tuinenburg, J.C.; Koning, G. & Seppenwoolde, Y. (2006). Is there an effect of flat-panel-
based imaging systems on quantitative coronary and vascular angiography?
Catheter Cardiovasc Interv 68:561-566.
Tuinenburg, J.C.; Koning, G. & Rareş, A. (2011). Dedicated bifurcation analysis: basic
principles. Int J Cardiovasc Imaging 27:167-74.
Van der Zwet, P.M. & Reiber, J.H. (1994) A new approach for the quantification of complex
lesion morphology: the gradient field transform; basic principles and validation
results. J Am Coll Cardiol 24:216–224.
Van Herck, P.L.; Gavit, L. & Gorissen, P. (2004). Quantitative coronary arteriography on
digital flat-panel system. Catheter Cardiovasc Interv 63:192–200.
Wellnhofer, E.; Wahle, A. & Mugaragu, I. (1999). Validation of an accurate method for three-
dimensional reconstruction and quantitative assessment of volumes, lengths and
diameters of coronary vascular branches and segments from biplane angiographic
projections. Int J Card Imaging 15:339–353 discussion 355–356.
Yong, A.S.; Ng, A.C. & Brieger, D. (2011). Three-dimensional and two-dimensional
quantitative coronary angiography, and their prediction of reduced fractional flow
reserve. Eur Heart J 32:345-53.
15

Summarized Coronary Artery Caliber


and Left Ventricle Mass for Scoring
of Cardiac Ischemia: Diagnostic
and Prognostic Value
Edvardas Vaicekavicius
Clinic of Radiology, Lithuanian University of Health Sciences
Lithuania

1. Introduction
1.1 Importance of methods for scoring coronary angiograms to reflect extent and
severity of CAD
Despite advances in modern medicine, the early detection of patients with obstructive
coronary lesions remains an unresolved problem due to frequently occurring atypical,
painless, and asymptomatic clinical variants of ischemic heart disease. For example, 80 to
90% of sudden cardiac deaths occur with a background of significant coronary artery
disease (Zipes, 1998) and sudden death is often the first presentation of the cardiac
disease. It is estimated that sudden death occurs in 33% to 70% of cardiac disease patients
(Davies, 2000). In addition, an acute myocardial infarction (MI) usually develops in
patients with multi-vessel coronary disease (Goldstein et al., 2000), while angiographic
studies have shown that an acute MI may develop in coronary segments having only
slight or moderate stenosis. It is well known that a ruptured atheromatous plaque in the
coronary artery can initiate intracoronary thrombosis and occlusion with the subsequent
development of an acute MI. However, plaque rupture with platelet activation and
thrombus formation are more recognized as the key events in the pathogenesis of acute
coronary syndrome than are the extent and severity of the coronary artery disease (CAD).
Additionally, the symptoms of myocardial ischemia may not be caused by obstructive
lesions in the coronary arteries at all. Myocardial ischemia may occur not only due to
decreased blood flow through epicardial coronary arteries, but also because of the
increased oxygen demand in patients with myocardial hypertrophy or hyperfunction.
Myocardial hypertrophy may be the cause of cardiac arrhythmias due to coronary
insufficiency in hypertensive patients. However, even in the absence of obstructive CAD,
hypertensive patients frequently have angina or may show electrocardiographic
abnormalities suggestive of myocardial ischemia due to coronary insufficiency (Vogt et
al., 1992).
In this era of great achievements in cardiology, we are still in need of a convenient and
informative coronary arteries analysis system that would allow us to quantitatively estimate
the size and severity of atherosclerotic CAD and myocardial ischemia burden.
274 Advances in the Diagnosis of Coronary Atherosclerosis

1.2 Consecutive steps in development of coronary scoring methods


Initially, the purpose of creating methods to score coronary angiograms was to assess the
severity of the underlying CAD and to determine whether the severity of angina and the
degree of altered left ventricular function were closely correlated with the severity and
extent of the underlying CAD. Multiple methods for evaluation and scoring of the coronary
angiograms were created. However, there was no difference in the frequency of angina
when the patients with the lowest coronary score were compared to patients with the
highest coronary score (Leaman et al., 1981). Clinically, it is possible to have severe angina
with one-vessel CAD or moderate angina with severe three-vessel CAD. Likewise, it is
possible to have well-preserved left ventricular function with severe three-vessel CAD as
well as to have severe left ventricular dysfunction with only one-vessel CAD.
Later, the coronary scores were used for risk stratification, because it was determined that
the prognosis and outcomes in patients were in large part dependent on the extent (one-,
two-, or three-vessel) and the severity of CAD (Ringqvist et al., 1983). This scoring system is
still used today. Sullivan et al., (1990) developed a new angiographic scoring system for the
extent of coronary disease (extent score). Therefore, the coronary scores seemed to be
essential for the selection of a tailored therapeutic strategy. Subsequently, the coronary
scores were used for the investigation of the relationship between the coronary lesion scores
and the levels of serum lipoproteins (Hamsten et al., 1986; Jenkins et al., 1978). Eventually, a
non-invasive coronary calcification scoring system became popular among investigators
because coronary artery calcification strongly correlated with the severity of coronary
atherosclerosis and the probability of obstructive disease (Kwon et al., 2011). The
development of an invasive coronary scoring system would have to be much more effective
in diagnosing and optimizing the treatment strategy, due to the fact that the non-invasive
methods could not reflect the extent and severity of CAD.
In order to increase the value of the coronary scores in the selection of coronary patients for
angioplasty, the American College of Cardiology/American Heart Association (ACC/AHA)
in 1988 presented a system of classification of coronary lesions to predict the success and
complications of balloon angioplasty (Krone et al., 2000). Although, the lesion type (A, B, or
C) was not predictive of complications, specific lesion morphologies were predictive of
adverse events and device use (Zaacks et al., 1998). Additionally, the type of lesion can
impact the outcome of coronary bypass operations (Graham et al., 1999; Jalal, 2007). Thus, to
choose an optimal revascularization strategy for patients with three-vessel and/or left-main-
stem disease, there is one important requirement: to quantify the complexity of CAD while
taking into account not only the number of significant lesions and their location, but also the
complexity of each lesion independently. For this purpose, the SYNTAX scoring system was
created (Garg et al., 2010; Sianos et al., 2005) The SYNTAX score was developed in order to
characterize the coronary vasculature in terms of the number, functional impact, location,
and complexity of the lesions.
However, in the myocardial infarction reperfusion era, it has become increasingly
apparent that clinical outcomes are not only associated with coronary flow in the
epicardial arteries but also with the quality of myocardial reperfusion. Quantitative
angiographic indexes of coronary artery blood flow -- TIMI (thrombolysis in myocardial
infarction) flow grades, corrected TIMI frame count, and TIMI myocardial perfusion
grade -- gave new insights into the pathophysiology of acute coronary syndromes (Gibson
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 275

et al., 1999, 2000, 2002; Korosoglou et al., 2007; Kunadian et al., 2009). Until recently, it
was assumed that the blood flow in the non-culprit arteries in patients with acute
coronary syndromes was normal. However, according to the data of Gibson et al., (2000)
the coronary flow in the non-involved arteries in acute MI was, in fact, 40% slower than
normal. It was also noted that the slower flow throughout all three arteries in acute MI
could be associated not only with the extensive damage of microvasculature circulation
but also might be the result of more extensive damage of non-culprit epicardial arteries.
Greater lesion complexity might also be associated with poorer epicardial and myocardial
perfusion.

1.3 The new challenges for coronary scoring


Because of these unresolved issues, it is imperative to extend the scope of investigations to
include the impact of multi-vessel lesions in epicardial coronary arteries in order to
elucidate the pathogenetic mechanism and prognosis of CAD. Therefore, we have created a
completely new scoring system for the quantification of coronarography and
ventriculography data. Using SYNTAX and other recent angiographic scoring methods, we
often have no opportunity to assess the impact of myocardial mass and function on the
myocardial ischemia and anginal severity. Hypertensive left ventricular hypertrophy is a
powerful predictor of coronary events (Ghali et al., 1992; Koren et al., 1991) and develops
due to impaired coronary blood flow autoregulation, decreased coronary reserve, increased
minimal coronary vascular resistance, and carries an increased risk of cardiac death in the
presence of coronary occlusion (Vogt et al., 1992). These events can be primarily dependent
on the size and severity of the coronary lesions and on the functional and morphological
status of the myocardium which requires a greater amount of blood for its nutrition. This
new angiographic scoring system, which assesses the extent of coronary disease, myocardial
mass, hypertrophy, and function, is simple to perform and it correlates better with the
clinical symptoms of CAD.

2. Methods and patients


2.1 Different technical solutions in calculation of coronary scores
The exact quantification of total coronary artery damage is very complicated in clinical
practice. Several different approaches have been proposed to determine the total coronary
score: 1) the number of vessels diseased, 2) the number of proximal arterial segments
diseased, 3) the proximal arterial segments score (Ringqvist et al., 1983), 4) the Friesinger
index (Friesinger et al., 1970), 5) the modified Gensini index (Gensini et al., 1971), 6) the
National heart and chest hospital (NHCH) index (Ringqvist et al., 1983), and 7) other
methods. The most common problems with these methods were: 1) the interpretational
differences in the estimation of the percentage of coronary artery narrowing due to
differences in the selection of optimal reference diameter (Brown et al., 1977; Dodge et al.,
1988), 2) the difficulties in the accurate assessment of the true hemodynamic value for each
narrowing, 3) the difficulties in angiographic quantification of diffuse CAD (Graham et al.,
1999; Jalal, 2007), and 4) the differences in techniques of measurements (Kalbfleisch et al.,
1990). The hemodynamic value used by some authors is set only by the degree of
narrowing. By other authors, the severity of luminal narrowing is weighted according to the
276 Advances in the Diagnosis of Coronary Atherosclerosis

usual flow to the left ventricle in each coronary vessel (Leaman et al., 1981) and according to
the flow to muscle mass rather than to the actual volume of flow (Brandt et al., 1977;
Gensini, 1983). However, it is difficult to accurately assess the coronary artery blood supply
zones due to variable coronary artery dominance and multiple branching options. Still some
authors tend to offset the impact of longevity of stenosis (Dodge et al., 1992) on myocardial
blood flow.
However, the determined angiographic CAD scores in most cases had a poor correlation
with the severity of angina pectoris and myocardial function. Therefore, the development of
new coronary scoring methods needs a different approach from those which have been
previously adopted for the accounting of coronary obstructions only.

2.2 Technique for determination of summarized caliber of coronary arteries and left
ventricle mass
It is well established that the development and progression of CAD involves three
processes: the reduction of the total caliber of coronary arteries, the increase in myocardium
weight, and the deterioration of left ventricular pump function. Therefore, we have sought
to create a specific coronary lesion scoring system which would avoid the subjectivity in the
determination of degree of stenosis and in the selection of normal reference diameter which
is highly variable and dependant on multiple variants in coronary dominance and
branching.
The severity of the coronary lesion was expressed by the sum of residual calibers of all
coronary arteries in the selected level. The extent of the coronary lesion was expressed by
the calculation of the summarized caliber of three levels (Fig. 1). The narrowest diameters of
the selected segments were used to calculate the coronary caliber expressed by coronary
square area (CSA = 0.785 × D2). The assessment of the SCCA at the first coronary branching
level included the sum of the cross-sectional areas at the narrowest points of the 1st and 5th
segments, at the second level – the areas of the 2nd, 6th, and 11th segments, and at the third
level – the areas of the 3rd, 7th , 9th, 12th , and 13th segments. The average of the arterial
calibers at each level was considered to be the SCCA.
The computer-assisted calculating algorithm for SCCA is very simple, and coronary
diameters can be measured using a quantitative analysis program readily available in
almost every modern angiographic unit. We measured the narrowest diameters of each
coronary segment in at least two projections of video images. In cases of uncommon
variants of coronary dominance or branching, we also measured and summed all segments
of the trunk below its second bifurcation.
Contrasting of the left ventricle was performed while the patient was taking a deep breath at
the right anterior oblique 30˚ body position. We performed a quantitative analysis of the
ventriculographic data using the radial method (Fig. 2). We computed the end-systolic and
diastolic volumes and estimated the myocardial mass by determining the wall thickness of
the left ventricle. The end-systolic and end-diastolic volumes were calculated using the
equation for spherical formations:

V = 4/3 π Ra3 × К (1)


In the equation above, we used the following abbreviations: V – systolic or diastolic volume,
Ra – average systolic or diastolic radius (cm), and K – a factor of correction.
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 277

Fig. 1. A scheme of 15 segments of the coronary tree proposed by the American Heart
Association which was used to calculate the summarized caliber of coronary arteries
(SCCA). The cross-sectional areas in the narrowest points of the segments at each level were
added together to represent the coronary caliber at one of the three coronary tree levels:
above the first bifurcation, below the first bifurcation, or below the second bifurcation of the
right and left coronary arteries. The average of the arterial calibers at the three levels was
considered to be the average SCCA.

Fig. 2. Radial scheme used in the quantitative analysis of ventriculographic data: A --


superposed end-systolic and end-diastolic profiles of the left ventricle with the α angle of
apex rotation; B -- superposed end-systolic and end-diastolic contours after the correction of
apex rotation; C –- a scheme of radial delineation; we used the midpoint of the long axis of
left ventricle (in diastole) to intersect the radial lines.
278 Advances in the Diagnosis of Coronary Atherosclerosis

We calculated the left ventricular mass using the thickness (h) of the anterior wall of the left
ventricle using the following equation:

LVM = (4/3 π × (h + Ra) 3 × K - 4/3 π Ra3 × K) × 1.05 (2)


In the equation above, we used the following abbreviations: LVM -- left ventricular mass, h
–- thickness of the anterior wall of left ventricle (cm), Ra --average systolic or diastolic radius
(cm), K – a factor of correction, and 1.05 – a comparative weight of the myocardium (g/cm3).
We measured the thickness of the anterior wall of the left ventricle at the midpoint between
the aortic valve and the apex.

2.3 Creation of the angiographic characteristics of myocardial supply


2.3.1 Angiographic coronary index
Initially, the SCCA was indexed to the supplied left ventricular mass. This parameter was
intended for the expression of the morphologic adequacy between the coronary artery tree
and the supplied myocardial mass. This diagnostic criterion was entitled angiographic
coronary index (ACI) and was calculated according the formula:

ACI = (SCCA/LVM) × 10 (3)


In the equation above, we used the following abbreviations: ACI -- angiographic coronary
index, SCCA -- summarized caliber of coronary arteries (mm2), LVM – mass of the left
ventricle (g), and 10 -- normalization factor used to reduce the average value of the index to 1.
During the post-mortem examinations, we performed a post-mortem coronary angiography,
an excision of coronary arteries by cutting them in intervals of 10 mm to analyze their
luminal square areas by planimetry. An autopsy of both ventricles was performed with the
separation of its masses with and without fatty tissues. We found that the patients without
any cardiovascular disease had the SCCA-to-LVM ratio approximately equal to 1. The ratio
was also almost the same as in the control group of patients who had no CAD symptoms.

2.3.2 Angiographic anginal index


Myocardial quantity (mass) does not represent the quality of myocardial tissue nor its
viability or its demand for arterial supply; instead, the greater myocardial mass usually
represents a lower functional status. To represent the functional status of the myocardium,
we used a ratio between the angiographic coronary index (ACI) and the ejection fraction of
the left ventricle (LVEF). We named the ratio an anginal index, because we noticed a very
prominent negative correlation between this index and the intensity of angina. However,
there was no correlation between the ACI and LVEF.
When the ACI exceeded 1.0 and the ACI was higher than the LVEF, the inadequacy between
the blood supply and the demand was theoretically absent. In contrast, when the ACI was
less than the LVEF, the inadequacy was present. The angiographic anginal index in these
cases was less than 1.0. Therefore, the angiographic anginal index allowed us to express
quantitatively the favorable conditions for myocardial ischemia.

2.3.3 Angiographic prognostic index


The complete characterization of the myocardial perfusion state and its prognostic impact
on the follow-up results using only the ACI-to-LVEF ratio was not possible. For example,
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 279

in patients with decreased SCCA and low LVEF (due to the presence of post-infarction
myocardial scars) the ratio did not show the inadequacy between the blood supply and
demand and masked the poor prognosis for these patients. Both the diminished SCCA
and the lowered LVEF may equally increase risk in patients. Therefore, the compound of
ACI and LVEF was labeled angiographic prognostic index (API). It was calculated using
this equation:

API = ACI + LVEF = (SCCA / LVM) + LVEF (4)


In the equation above, the API stands for the angiographic risk index, ACI -- angiographic
coronary index, SCCA -- summarized caliber of coronary arteries (mm2), LVM – mass of the
left ventricle (g), and LVEF – ejection fraction of the left ventricle.

2.4 Patient groups


In this way, we have quantitatively analyzed coronarographic and ventriculographic data in
403 patients with different clinical forms of ischemic heart disease. All patients were divided
into groups according to the size and severity of coronary obstructions and the left
ventricular mass. There were 59 (14.6%) patients with normal coronary arteries, 165 (40.9%)
patients with arteries narrowed up to 50% of diameter, and 179 (44.4%) patients with lesions
which obstructed more than 50% of the coronary artery diameter. Among patients with
coronary narrowing of up to 50%, 88 (24.8%) patients had a normal ventricular weight and
77 (19.1%) had an increased weight of the left ventricle. The patients with increased left
myocardial weight were divided into two subgroups: the cardiomyopathy (obstructive and
hypertrophic) subgroup and the secondary hypertrophy due to hypertension subgroup. The
majority of patients (82 or 45.8%) with obstructions of more than 50% had coronary disease
of a single branch, 67 (37.4%) patients had disease in two branches, and the remaining 30
(16.7%) patients had lesions in all three branches. Twenty six (14.5%) patients had left main
coronary artery disease. The average age of patients was 47.8 ± 0.4 years. The dominant
gender was male (95.5%). Additionally, patients were divided into groups according to
clinical diagnosis (stable angina pectoris, post-infarction angina, hypertensive cardiopathy,
and hypertrophic cardiomyopathy). The follow–up results after 2 to 7 years were analyzed
in 136 patients with CAD (52 underwent surgery, and 84 were treated conservatively).
Thirteen of these patients died after surgical intervention, and nineteen died after
conservative treatment.
We performed 176 necropsy examinations with postmortem coronarography and
epicardial excision of the coronary trunks. The trunks were cut in slices for morphologic
analysis and the myocardium of the right and left ventricles was separated into three
parts according to the supply zones of the anterior descending artery, the circumflex
branch of the left coronary artery, and the right coronary artery. The majority (n = 146) of
these patients died from CAD: 70 patients experienced sudden death and the rest died
from different complications of CAD. Three control groups were formed from the
remaining 30 patients who died from other, non-cardiac diseases: the 1st control group
was comprised of patients who died younger than age 50, the second control group was
comprised of patients who died older than age 50, and the third control group included
patients of any age who died having signs of cardiac hypertrophy without any
obstructions in the coronary arteries.
280 Advances in the Diagnosis of Coronary Atherosclerosis

2.4.1 Statistical analysis


The study data was processed using STATISTICA 5 and SPSS 10 (Statistical Package for
Social Science) software. All data were presented as mean values ± standard deviations. The
following statistical methods of analysis were used: paired t-test for differences in groups, t-
test for differences between two groups, ANOVA for differences between multiple groups,
and chi-square (χ2) – for categorical data. A p-value less than 0.05 was regarded as
statistically significant.

3. Results
3.1 Calculation of angiographic coronary index using autopsy data
The necropsy study consisted of the following parts: 1) performance of the post-mortem
coronary angiography (Fig. 3A); 2) the longitudinal excision of contrasted coronary artery
trunks from the myocardial tissue and cutting them into 10 mm-long pieces for microscopic
analysis, and 3) the division of the myocardial mass of both ventricles (with and without
fatty tissues) into three parts according to the supply regions of the right coronary artery,
the circumflex branch and the anterior descending artery (Fig. 3B).

A B

Fig. 3. The anteroposterior view of postmortem coronary angiography (A). Myocardial


cutting into parts differing according to blood supply regions: in the upper left position is
the part supplied by the circumflex branch, in the upper right is the part supplied by
anterior descending artery, and in the bottom is the part supplied by the dominant right
coronary artery (B).
The coronary arteries were contrasted with 40% solution of BaSO4. The contrast material
was injected using 120 mmHg pressure through a special cannula inserted and fixed into
the orifice of the right and left coronary arteries (Fig. 3). The coronarograms were made
after the stagnation of contrast material. The measurements of the coronary arteries were
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 281

performed using the methodology for the determination of SCCA. The second step was
supplemented by weighing the different parts of the myocardium, by calculation of the
coronary caliber, and by the assessment of the three separate ACIs for each coronary
artery.
The examination of the arterial cuts revealed the diffuse nature of the obstructive lesions in
patients who died suddenly or died after complications of chronic heart disease. In the
control group of younger patients, the slight narrowing (less than 50%) covered only 5.9 -
8.1% of the coronary artery length. In the second control group of older patients, the same
lesions covered 9.8 to 13.3% of coronary artery length. However, in patients who died from
CAD, the total length of the obstructed segments covered from 73.6% to 91.6% of coronary
artery length; furthermore, one-half of the segments had less than 50% narrowing. This
means that the quantitative estimation of the degree of stenosis using the referential normal
adjacent segment for calculation may often be incorrect.
The assessment of the coronary artery diameter without the calculation of the degree of
stenosis is more objective. The SCCA in control patients was almost the same at each level
and there were no significant differences between the control groups. However, the
difference between each level SCCA of the two groups of died patients, from one side, and
of the three control groups, from other side, was noticed (Table 1). These findings are a
consequence of differences in the size, location, and severity of obstruction in CAD patients.
At the same time the decrease in SCCA with the simultaneous increase in myocardial
weight was fixed in patients who died from sudden coronary death or from CAD
complications (Table 2).

No. SCCA of different levels, mm2


Patient groups of
1st level 2nd level 3rd level
pts.
1st control group
12 27.1 ± 5.2 29.1 ± 1.5 24.8 ± 4.2
(age 34.2 ± 10.2 yrs.)
2nd control group
9 25.2 ± 5.4 25.6 ± 6.0 25.0 ± 4.6
(age 67.5 ± 9.7 yrs.)
3rd control group
7 23.6 ± 7.2 25.6 ± 5.6 24.3 ± 4.5
(LV hypertrophy)
4th group (sudden
14 14.9 ± 6.8 7.58 ± 3.3 11.7 ± 4.2
coronary death)
5th group (death from
17 12.9 ± 7.4 10.3 ± 4.0 7.8 ± 4.7
CAD complications)
Table 1. The summarized caliber of coronary arteries of three levels in different groups of
died patients.
The post-mortem assessment of the ACI in the first and second control groups has shown
that the value of the ACI slightly exceeded 1. Additional analysis showed that the ACI of the
right coronary artery was lower than the ACI of the left coronary artery (р < 0.03). This
distinction could be caused by functional peculiarities of both ventricles. Because the SCCA
was almost the same in all three groups, the diminished ACI in the third group was
associated with the increased weight of the myocardium. However, the SCCA in patients of
282 Advances in the Diagnosis of Coronary Atherosclerosis

the fourth and the fifth groups with CAD was significantly smaller, and the ACI was
consequently low, also.

SCCA, ACI, and myocardial mass


No.
Myocardial
Patient groups of Average SCCA,
ACI mass without
pts. mm2
lipid tissue, g
1st control group
12 27.2 ± 3.9 1.41 ± 3.90 226.4 ± 11.0
(age 34.2 ± 10.2 yrs.)
2nd control group
9 25.3 ± 4.4 1.42 ± 0.16 209.9 ± 12.2
(age 67.5 ± 9.7 yrs.)
3rd control group
7 25.0 ± 4.4 0.84 ± 0.06 337.8 ± 4.90
(LV hypertrophy)
4th group (sudden
14 11.1 ± 3.2 0.14 ± 0.04 306.0 ± 17.6
coronary death)
5th group (death from
17 9.9 ± 4.5 0.22 ± 0.04 305.1 ± 12.7
CAD complications)
Table 2. The SCCA, ACI, and myocardial mass in different groups of deceased patients.
Six sudden death patients in the fourth group had no obvious segmental obstructions in
their coronary arteries; however, the ACI was significantly lower (0.62 ± 0.01) than the
control values (р < 0.001). The lowered ACI was caused by increased left ventricular weight
and lowered SCCA, compared to the control groups. The detailed analysis of the
myocardium in these patients did not revealed any signs of myocardial infarction. We may
assume that the relative insufficiency could be the starting mechanism for the expressed
myocardial ischemia and for the possible life threatening arrhythmia. This background
factor of inadequacy between the SCCA and the myocardial mass could be a cause of the
sudden coronary death.

3.2 Assessment of angiographic characteristics of myocardial supply (ACMS) in


clinical setting
Various authors have expressed difficulties in creating an unbiased angiographic coronary
scoring system intended for researchers as well as for clinical practice. In particular, all the
diagnostic and prognostic capabilities of angiographic coronary scoring remain
undetermined.

3.2.1 Difficulties in assessment of angiographic coronary scores associated with the


anatomical variability of coronary dominance and branching
Because only three types of anatomic variants of coronary dominance are commonly used in
previous scoring systems and all possible coronary branching types are not taken into
consideration, the hemodynamic value of each stenosis might be calculated inaccurately.
We have analyzed the impact of coronary dominance on the diameters of all fifteen
coronary segments while using not three but six types of coronary dominance. The six types
were determined using the ratio between the myocardial mass supplied by the right and the
myocardial mass supplied by the left coronary arteries. Using a topographic scheme shown
in Fig. 4, we have calculated the ratio by delineating myocardial zones supplied by the right
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 283

and left coronary arteries. Coronarograms performed in the left and right anterior oblique
30˚ views are shown in the upper part of the figure. The views are most informative in
understanding the branching peculiarities of both coronary arteries supplying the posterior
and inferior walls of the left ventricle. A percept used to calculate the amount of myocardial
tissue supplied by the right and left coronary arteries is shown in the lower part of the
picture. This approach was chosen according the information obtained from post-mortem
studies. The whole myocardial tissue (mass) was assigned 20 conceptual mass units. Five
units were assigned to the right ventricle, another five to the septum (3 units to the anterior
and 2 units to the posterior septum), and the remaining 10 units were assigned to the mass
of the left ventricle divided into ten equal parts.

Fig. 4. Topographic schemes reflecting the delineation and calculation of myocardial


zones and mass supplied by the right and left coronary arteries. The lines divide
myocardial tissue of the left ventricle into 10 zones (units), each of similar mass. Using the
same principle, the right ventricle was assigned 5 units, the anterior septum -- 3 units, and
the posterior septum -- 2 units.
Using this technique we could quantitatively define all possible variants of coronary blood
supply to the right and left myocardial masses. To indicate the type of coronary dominance
we used the ratio between myocardial masses supplied by the right and left coronary
arteries. We used the R:L notation (where R and L correspond to the number of mass units
supplied by the right and left coronary arteries). For example, the 5:15 ratio would indicate
that 5 units were assigned to the right, and 15 units to the left myocardial supply zone. We
have encountered the six most common variants of coronary dominance -- 5:15 (potentially,
284 Advances in the Diagnosis of Coronary Atherosclerosis

the variant of the left dominance), 6:14; 7:13; 8:12; 9:11, and 10:10 (potentially, the variant of
the right dominance).

Fig. 5. The quantitative expression of coronary dominance: A, 12:8; B, 9:11; C, 6:14; D, 4:16.
The usage of the right-to-left myocardial mass ratio allowed us to define the coronary
dominance variants more precisely (Fig. 5). Dividing control patients into the six groups
(according to the most common coronary dominance variants) allowed us to obtain
significant differences in segment diameters of the circumflex branch of the left coronary
artery and the right coronary artery between all groups. However, there was no statistical
difference between all groups according to the segments of the left anterior descending
artery, because the caliber of these segments is more stable with respect to the variations in
coronary dominance.
We have identified the four most frequent variants of coronary branching (Fig. 6) while
using these topographic schemes for a more thorough analysis of the coronary supply
zones. It is interesting to note that the following four variants of coronary branching had
the exact same ratio of coronary dominance determined by the myocardial supply zones.
However, the correlation between the caliber of the coronary segments and the size of their
myocardial supply zones was found to be very high (г = 0.96) despite the variability of
coronary dominance and branching. This finding shows the fundamental importance of
assessing the contiguous segments to estimate the real degree of stenosis, because diameters
of adjacent arterial segments usually visible in coronarograms may be diminished because
of the diffuse obstructive changes in adjacent coronary arteries, and it becomes very difficult
to decide whether the particular segment is damaged or not. Therefore, the assessment of
the coronary lesion score using SCCA appears to be more objective; it does not require
consideration of the coronary dominance and branching. At the same time, the location and
degree of atherosclerotic obstructions may be detected automatically by measuring the
diameter of the narrowest point in every segment at several levels. Therefore, to increase the
accuracy of coronary scoring, it is important to calculate the SCCA in a higher number of
levels.
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 285

Fig. 6. The most frequent variants of coronary branching: A, common (6:14); B, left anterior
descending dominance (6:14); C, trifurcation (6:14); and D, marginal (6:14).

3.2.2 Correlation between summarized caliber of coronary arteries and different


angiographic coronary scores
We have analyzed the correlation between the data obtained by SCCA and by other
coronary scores. The diagnostic features of the Friesinger, Leaman, and Gensini scores or
indexes dedicated for the quantitative expression of the extent and severity of CAD were
compared with each other and with SCCA.
The Friesinger index ranges from 0 to 15. Each of the three main coronary arteries is scored
separately from zero to five. The scores are: 0 -- no arteriographic abnormalities; 1 – a trivial
luminal narrowing less than 29%; 2 – a localized 30 to 68% luminal narrowing; 3 -- multiple
30 to 68% luminal narrowing; 4 -- 69 to 100% luminal narrowing without 100% occlusion of
proximal segments; and 5 -- total obstruction of a proximal segment.
The Leaman index has another weighting system. If the vessel was totally occluded, the
coronary artery segment value was multiplied by 5.0. If there was a 90 to 99% occlusion, it
was multiplied by 3.0. For a 70 to 89% obstruction, it was multiplied by 1.0. The segmental
scores were then added to derive the total coronary score.
The modified Gensini index assigns a heavier weight to the more severe luminal narrowing
(Gensini, 1983). Weights are also assigned to each segment depending on vessel size and
importance; segments serving larger regions of myocardium are more heavily weighted. For
example, the left main coronary artery receives the heaviest weight. Each arterial segment is
weighted by a value from 0.5 to 5.0. (The weight for segments 1 through 4 is changed to 0.5
if the system is left dominant.) The stenosis is weighted from 2 to 64. The product of these
two weights is the total weight for each arterial segment. The modified Gensini index is the
sum of the total weights for each segment. For each segment the two weights are multiplied.
The sum of the products is the modified Gensini score.
So, we have compared fundamentally different angiographic coronary scoring methods
taking into account: 1) the degree of stenosis regarding the diameters of adjacent segments,
286 Advances in the Diagnosis of Coronary Atherosclerosis

2) the degree (weight) of stenosis for each segment regarding the three types of coronary
dominancy, and 3) the average SCCA. The comparative analysis of different scoring
methods showed that the lowest correlation was observed between the methods that were
based on different coronary scoring principles. For example, Friesinger index has only
moderate correlation with ACI (r = -0.45). The correlation between Gensini index and ACI
was only insignificantly better (r = -0.54). However, the correlation between Friesinger and
Gensini indexes was high (r = 0.84).

3.3 Value of angiographic characteristics of myocardial supply in diagnostic


improvements
Primarily, we investigated the relationship between angiographic characteristics of
myocardial supply (ACMS), on the one hand, and clinical symptoms of myocardial ischemia
and clinical forms of ischemic heart disease, on the other hand. For the expression of
myocardial supply features by angiographic characteristics, we have chosen what we
consider the three most important summarized indices of all angiographic indices, namely,
the angiographic coronary index (ACI), the angiographic anginal index (AAI), and the
angiographic prognostic index (API).

3.3.1 Interrelation between clinical symptoms of CAD and the angiographic


characteristics of myocardial supply
The dependence of ACMS from the functional class of angina pectoris determined according
to the classification of Canadian Cardiovascular Society is shown in Table 3. The data show
that there is a very well defined relationship between all ACMS and the clinical severity of
anginal symptoms. In addition, we have analyzed the relationship between the ACMS and
the character of anginal pain (Table 4). Patients were divided into 3 groups: the 1st group
consisted of patients with atypical prolonged anginal pain, the 2nd group -- of patients with
typical anginal symptoms, and the 3rd group -- of patients who had typical pain but after an
effective treatment of acute MI the anginal symptoms disappeared.

Class of ACI
No. of SCCA, AAI
Angina (SCCA / LV mass) API (ACI+EF)
pts. mm2 (ACI /LVEF)
Pectoris
1˚ 49 23.4 ±1.0 0.68 ± 0.03 1.0 ± 0.06 0.78 ± 0.07
2˚ 21 18.0 ± 1.1 0.49 ± 0.04 0.8 ± 0.08 0.45 ± 0.08
3˚ 22 13.2 ± 0.7* 0.39 ± 0.05 0.6 ± 0.03 0.23.9 ± 0.14
1 vs. 2 0.001 0.001 0.019 0.006
P 1 vs. 3 0.001 0.013 0.007 0.007
2 vs. 3 0.005 0.009 0.05 0.05
Table 3. ACMS in patients with different functional class of angina pectoris.
All angiographic characteristics were worse in patients with typical angina (2nd group)
compared to patients with atypical chest pain (1st group). Similar significant differences
were discovered between the 1st and 3rd groups of patients who did not have the post-
infarction angina. The patients who did not have the post–infarction angina also had worse
average SCCA, ACI, and anginal index. It is interesting that there were no differences in
ACMS between patients in the 2nd (typical angina) and 3rd groups (no post-MI angina).
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 287

Average
Character of No. of ACI AAI API
SCCA,
anginal pain pts. (SCCA / LV mass) (ACI /LVEF) (ACI+EF)
mm2
0.71 ±
1˚ atypical AP 49 27.3 ± 1.2 71.1 ± 4.1 1.0 ± 0.05
0.07
0.48 ±
2˚ typical AP 21 20.8 ± 1.2 57.2 ± 2.9 0.83 ± 0.06
0.05
3˚ disappeared
22 22.9 ± 2.2 48.7 ± 4.4 1.33 ± 0.1 0.63 ± 0.1
post-MI AP
1 vs. 2 0.001 0.009 0.05 0.05
P 1 vs. 3 0.05 0.001 0.05
2 vs. 3 0.004
Table 4. Angiographic characteristics of myocardial supply in patients with different
character of anginal pain. The statistical significance of the differences in angiographic
characteristics between the three different characters of anginal pain is shown in the lower
part of the table.
However, the difference in the angiographic anginal index between these three groups
existed not because of the difference in SCCA but because of the difference in left-
ventricular ejection fraction that was evidently lower in post-MI patients. Furthermore,
there was no inadequacy between the diminished SCCA and the diminished left-ventricular
ejection fraction in post-infarction patients.

3.3.2 Interrelation between clinical forms of coronary artery disease and angiographic
characteristics of myocardial supply
We have studied the levels of ACMS in patients divided into groups according to the
different clinical forms of ischemic heart disease. The values of different ACMS obtained
from patients with different clinical diagnoses of ischemic heart disease are shown in Table
5. The SCCA was highest in control patients and lowest in patients with multivessel CAD.
The SCCA of all patients groups with CAD was lower compared with the groups without
CAD. This consistent pattern was observed in the comparison of patients according to ACI,
except that the ACI was the same in patients with one vessel disease and in patients with
myocardial hypertrophy. The same regularity was noted between groups compared
according to AAI and API. The anginal syndrome was expressed mostly in patients with
multi-vessel disease. The AAI and API were equally expressed between patients with one-
vessel disease and patients with myocardial hypertrophy.
The various ACMS in patients with different clinical forms of myocardial infarction are
shown in table 6. The 1st group included patients with myocardial infarction without ST
segment elevation (NSTEMI). The 2nd group included patients with ST elevation myocardial
infarction (STEMI).
After comparing both groups of patients, we noticed that NSTEMI and STEMI were
significantly different according to AAI and API only. The SCCA and ACI differed
insignificantly. The API was worse in NSTEMI patients. Similar findings confirm the studies
of other authors.
288 Advances in the Diagnosis of Coronary Atherosclerosis

No. of ACI AAI


Diagnosis SCCA, mm2 API (ACI+EF)
pts. (SCCA / LV mass) (ACI /LVEF)
Stable AP in
patients with
101 18.9 ± 0.661 57.2 ± 2.322 0.82 ± 0.043 128.4 ± 2.754
one-vessel
disease
Stable AP in
patients with
34 12.6 ± 1.40 30.3 ± 3.47 0.50 ± 0.07 93.7 ± 5.06
multi-vessel
disease
Stable post-
96 18.7 ± 0.831 49.2 ± 2.41 1.04 ± 0.07 101.9 ± 3.40
infarction AP
Stable AP with
myocardial
hypertrophy & 110 23.7 ± 0.64 56.0 ± 1.362 0.82 ± 0.0243 127.2 ± 2.094
insignificant
CAD
Controls
without CAD 35 32.8 ± 1.23 95.2 ± 4.44 1.5 ± 0.091 163.2 ± 6.0
and AP
Table 5. ACMS in different groups of patients with ischemic heart disease. Statistically
significant differences (P < 0.01) were found between all groups of patients, except the
groups shown in superscript.

MI in No. of SCCA, ACI AAI API


anamnesis pts. mm2 (SCCA / LV mass) (ACI /LVEF) (ACI+EF)
NSTEMI
21 18.1 ± 1.0 51.7 ± 3.2 0.93 ± 0.08 0.45 ± 0.05
(1st group)
STEMI
22 21.3 ± 1.1 55.1 ± 5.5 0.75 ± 0.04 0.79 ± 0.08
(2nd group)
P value 2 vs. 3 0.001 0.008

Table 6. ACMS in groups of patients with different clinical forms of myocardial infarction.

3.4 Evaluation of angiographic characteristics of myocardial supply in predicting of


outcomes
Death of cardiac origin has occurred in only 32 out of 136 patients analyzed during the
seven-year follow-up period. We did not notice any progression of disease after 3 to 7 years
in 40 patients with CAD who had a slightly lower ACI and left-ventricular ejection fraction
but normal prognostic index. We performed a detailed analysis of ACMS indices in patients
who died within three years (Table 7).
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 289

Myocardial
ACI
infarction No. of SCCA, AAI API
LVEF (SCCA /
in pts. mm2 (ACI /LVEF) (ACI+EF)
LV mass)
anamnesis
Death
during the 1.04 ±
7 20.7 ± 8.2 0.53 ± 0.03 0.51 ± 0.04 0.98 ± 0.03
3rd year 0.05
(1st group)
Death
during the 0.60 ±
5 22.7 ± 6.6 0.29 ± 0.03 0.31 ± 0.05 1.07 ± 0.05
2nd year 0.08
(2nd group)
Death
during the 1.03 ±
6 14.3 ± 0.1 0.67 ± 0.04 0.36 ± 0.06 0.53 ± 0.06
1st year 0.09
(3rd group)
1 vs. 2 0.001 0.05 0.001
P 1 vs. 3 0.001
2 vs. 3 0.001 0.001 0.01
Table 7. ACMS in patients who died within three years.
All 18 patients were divided into 3 groups: the 1st group (n = 7) consisted of patients who
died during the third year of investigation, the 2nd group (n = 5) of patients who died during
the second year of investigation, and the 3rd group (n = 6) of patients who died during the
first year of investigation. Patients who died during the first year after coronarography had
the most diminished average SCCA, but their left-ventricular ejection fraction was normal.
These patients usually experienced a sudden coronary death. However, the patients who
died later, during the 2nd year after investigation, had not only a lower ACI but also a lower
left-ventricular ejection fraction. They usually died from heart failure. It is interesting to
point out that these patients had a worse API and a normal AAI. They did not have typical
anginal symptoms. The patients with a lower ACI and a decreased LVEF lived slightly
longer despite the fact that they had clinical symptoms of heart failure but they were treated
more intensively. It is very important to emphasize that inadequacy between ACI and
ejection fraction, when left ventricular hypertrophy has developed, increases the risk for
sudden cardiac death. This is a consequence of expressed multi-vessel CAD in patients with
viable myocardial hypertrophy. In patients who survived three years, the left-ventricular
ejection fraction, ACI and API were better. Therefore, we believe that the survival and death
of patients with CAD can be better predicted with the help of ACMS. The product of both
angiographic criteria (the ACI expressing the supply adequacy to the myocardial mass and
the anginal index expressing the adequacy of supply to the functional requirements) could
be the most informative prognostic index of survival in CAD. The product of ACI and
ACI/EF could be named the angiographic outcome index (AOI). Mathematically, it can be
calculated using the following equation:
290 Advances in the Diagnosis of Coronary Atherosclerosis

AOI = ACI × (ACI / EF) = (SCCA / LVM) × (SCCA / LVM) / EF = SCCA2 / LVM2 × EF (5)
In the equation above the ACI stands for the angiographic coronary index, AOI –
angiographic outcome index, SCCA -- summarized caliber of coronary arteries (mm2), LVM
– mass of the left ventricle (g), and EF – ejection fraction of the left ventricle. The AOI was
statistically different in all three groups of patients who died during the 1st, 2nd, or 3rd year
after intervention. It was subsequently 0.1 ± 0.03, 0.33 ± 0.02, and 0.5 ± 0.02 (p < 0.009, p <
0.001, and p < 0.007).

4. Discussion
The quantification of coronary stenosis is an important practice in cardiology from both a
clinical and a research standpoint. In the recent past, there have been several imaging
methods which could differently characterize the size and severity of obstructive CAD:
coronary angiography, computed tomography, magnetic resonance imaging, and
intravascular ultrasound. Various techniques with differing degrees of sophistication were
used to measure the amount of luminal narrowing. Each method has provided new
opportunities for a more thorough understanding of the pathogenesis of ischemic heart
disease and variability of clinical picture. Angiography, performed with the use of digital
techniques, has been shown to have significant advantages over the above-listed techniques
due to its excellent reproducibility and accuracy (Funabaschi et al., 2003; Kalbfleisch et al.,
1990; Molloi et al., 2001). However, for the effective computerized assessment of coronary
lesion scores, we need to develop programs which can help to determine the size and
severity of CAD more accurately. To achieve this aim, the proposed SCCA assessment
system could be useful both theoretically and practically. Let us compare this innovative
system to other methods of analysis.
All scoring systems have been based on percentage of stenosis evaluation using the
“normal” diameter in the adjacent coronary segment. The validity of this reference is in
question because coronary atherosclerosis is often diffuse and the adjacent segment could
also be narrowed (Marcus et al., 1983; Seiler et al., 1992). In addition, serial segmental
narrowing representing a type of diffuse disease is not taken into account because of the
absence of a true normal reference segment. To assess the severity of diffuse CAD (with or
without segmental narrowing) it is necessary to know what the normal arterial lumen size
would be at each segment in the absence of coronary atherosclerosis. Various authors have
attempted to solve this problem in different ways (Kucher et al., 2001; Seiler et al., 1992).
Kucher et al. (2001) showed that in a population without cardiac disease, women have
smaller coronary artery size even after normalization for left ventricular mass. The existence
of a gender difference in coronary artery size remains controversial.
Our search for a “normal” diameter at each segment showed that the successful solution to
this problem is impossible due to high variability in coronary dominance and branching.
However, our experience and several other experimental and clinical reports (Dodge et al.,
1992; Roberts C & Roberts W, 1980; Seiler et al., 1992) have shown a direct relationship
between the coronary artery lumen size and the myocardial mass supplied by this artery.
Therefore, the variability in coronary diameter and its direct relationship to myocardial mass
have led us to propose a methodology which could be used to assess the degree and cause of
any imbalance between residual coronary diameter and the supplied myocardial mass.
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 291

An analysis of patients with intact coronary arteries and increased myocardial mass showed
that the SCCA can increase along with the degree of myocardial hypertrophy, especially if
this process takes place at a young age. This phenomenon does not disturb the balance
between SCCA and left ventricular mass. Therefore, the ratio remains normal. In cases of
myocardial hypertrophy in older populations where the process of atherosclerosis has
begun, the SCCA does not increase in conjunction with myocardial mass. This relative
discrepancy is well illustrated by the lowering of the angiographic coronary index. Similar
findings have been described by Marcus et al., (1983). Myocardial ischemia is frequently
observed in patients with cardiac hypertrophy even when the conduit coronary arteries are
normal (Fig. 7.2 and 7.3). Recent studies indicate that impaired coronary reserve in
hypertrophied hearts probably occurs because the growth of the coronary bed does not keep
pace with increase of cardiac mass (Fig. 7.4). The imbalance between vascular proliferation
and muscle growth is probably most severe when the cardiac hypertrophy is caused by
pressure overload or atherosclerotic infiltration (Fig. 7.5). These observations suggest that
abnormalities in the coronary microcirculation which accompany cardiac hypertrophy play
a significant role in the pathogenesis of the complications associated with cardiac
hypertrophy.
The diminishing ACI in people without blockage of epicardial coronary arteries usually is
associated with the onset of typical or atypical anginal symptoms. Syndrome X can be
associated with chest pain and chest discomfort in people who do not show signs of
obstruction in the larger coronary arteries. No one knows exactly what causes Syndrome X
and it is unlikely to have a single cause. Today, we speculate that Syndrome X may be
caused by microvascular dysfunction. A large majority of women have typical anginal
symptoms which are not associated with the presence of atherosclerotic plaques. Scientists
speculate that the blood vessels in these women are diffusely abnormal and smaller than in
males (Kucher et al., 2001). It is possible that the diminishing ACI is a causative factor in the
development of Syndrome X.
Additionally, the relationship between the angiographic coronary index and the functional
status of the left ventricle expressed by ejection fraction may specify the adequacy of
myocardial supply to myocardial functional demands. First, the inadequacy of myocardial
supply with the inadequacy of myocardial nutritional requirements may lead to the
occurrence of ischemic pain and its severity. The relationship between the angiographic
coronary index and the ejection fraction we named the angiographic anginal index, because
we found a strong negative correlation between this index and the intensity of angina.
Recent techniques have revealed new characteristics and expanded our understanding of
painless or silent myocardial ischemia. These techniques include treadmill exercise testing;
radioisotope techniques, including ejection fraction studies, stress thallium scintigraphy,
and tomographic imaging. These new noninvasive tests should be used to detect transient
ischemia, estimate its severity, and measure patient's risk for adverse coronary events
especially in patients with painless or silent myocardial ischemia (Selwyn, 1990). Usage of
anginal angiographic index in clinical practice can facilitate the selection of such high-risk
patients with silent ischemia. Patients with diabetes, chronic renal insufficiency and other
coexisting diseases have an increased prevalence of coronary artery disease with silent
myocardial ischemia, complex ventricular arrhythmias, atrial fibrillation, and left
ventricular hypertrophy (Das et al., 2006; Chipkin et al., 1987). These risk factors for
cardiovascular morbidity and mortality contribute to the increased incidence of
292 Advances in the Diagnosis of Coronary Atherosclerosis

cardiovascular morbidity and mortality seen in patients with the above mentioned
concomitant diseases (Selwyn, 1990). It is also important to stress that the energy needs of
the left ventricle are not always proportional to the ejection fraction. LVEF can be
permanently reduced due to post-infarction scar development, but the LVEF may
temporarily decrease due to inadequate blood supply to the heart muscle. This phenomenon
is known as myocardial hibernation (Camici et al., 2008). In both situations with the
disappearance of anginal pain the occurrence of symptomatic heart failure can manifest
with the clinical symptoms of ischemic cardiomyopathy (Felker et al., 2002).

Fig. 7. Changes occurring in the coronary angiogenesis during development of myocardial


hypertrophy. The size of the square indicates the presence of myocardial hypertrophy and
its degree. The number and size of vessels are shown in each shape: 1, normal number of
vessels in non-hypertrophied myocardium; 2 and 3, increased number of normal vessels in
hypertrophied myocardium; 4, the non-increased number of normal vessels in
hypertrophied myocardium; 5, increased number of atherosclerotic thickened-wall vessels
in hypertrophied myocardium; and 6, increased number of spastic vessels in hypertrophied
myocardium.
The diagnostic value of angiographic anginal index can be confirmed by the fact that during
the development of myocardial hibernation or ischemic cardiomyopathy anginal pain reduces
and can disappear completely. During the last decade, the existence of totally asymptomatic
myocardial ischemia has been conclusively established. We would like to emphasize that the
severity of angina depends upon the inadequacy between myocardial nutrition and functional
demand, and this was shown by the angiographic anginal index. This index reflecting the
adequacy between myocardial supply and demand cannot express the real degree of coronary
and myocardial damage, because there may be a supply-demand adequacy in patients with
equally expressed damage in the coronary arteries and the myocardium. Knowing the limited
prognostic capabilities of the angiographic anginal index, we have created the angiographic
outcome index for outcome prediction, and the angiographic prognostic indexes.
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 293

The prognostic capabilities of different coronary scores were confirmed by several authors
(Friesinger et al., 1970; Graham et al., 1999; Korosoglou et al., 2007; Ringqvist 1983). The
complex angiographic coronary index, anginal index, and left-ventricular ejection fraction
are more informative for predicting CAD outcomes. It was shown that only an increase in
LV mass in initially healthy persons with essential hypertension resulted in more
complications and deaths (Koren et al., 1991). Differently determined left ventricular
hypertrophy is an important prognostic marker in patients with or without coronary artery
disease (Ghali et al., 1992). It was shown that only reduced LVEF has a clear predictive value
in patients without known obstructive CAD. Reduced LVEF was related to sudden death
from the absolute presence of CAD as well as the increased number of vessels with
obstruction (Min et al., 2010).

4.1 Limitations
We have measured the lumen areas of different segments at their narrowest points on
coronarograms in this study. Very small measurements sometimes cannot be done without
interpretative errors; however, the margin of error is not high. The possibility of
underestimating the impact of a single expressed lesion on the diminishing SCCA could be a
limitation of this method. It is well known that such single-culprit lesions may cause the
development of acute myocardial infarction. However, the main goal of this quantitative
analysis system is to assess the total real size and severity of the epicardial coronary lesion
using the SCCA as a coronary score.

5. Conclusion
The most important advantage of this SCCA calculation system is that it provides an easier
and more objective assessment of an angiographic coronary lesion score in patients with a
wide spectrum of CAD, ranging from a minimal stenosis to extensive diffuse involvement of
the coronary arteries. The SCCA as a coronary lesion score is simple and easily interpreted;
it gives no reason for interpretative differences. First, the estimation of the average SCCA
does not require a precise assessment of stenosis degree, coronary dominance, or branching
peculiarities. Second, it provides the possibility to analyze the relationship between
coronary score and myocardial function what implies a correlation between myocardial
supply and demand. In addition, this quantitative analysis system can give more diagnostic
and prognostic information about the real mechanisms of myocardial ischemia, the
character of angina pectoris, and the possible risk for occurrence of life threatening
complications. Furthermore, the technique for interpretation of angiographic data and
computation of quantitative characteristics has been considerably facilitated.

6. Acknowledgment
I would like to thank the staff of the Department of Clinical Pathology, Lithuanian
University of Health Sciences for their valuable assistance in performing post-mortem
examinations.

7. References
Brandt, P.; Partridge, J. & Wattie, W. (1977). Coronary arteriography: method of presentation
of the arteriogram report and a scoring system. Clin Radiol, Vol. 28, pp. 361-365,
ISSN 0009-9260
294 Advances in the Diagnosis of Coronary Atherosclerosis

Brown, B.; Bolson, E.; Frimer, M. & Dodge, H. (1977). Quantitative coronary arteriography:
estimation of dimensions, hemodynamic resistance, and atheroma mass of
coronary artery lesions using the arteriogram and digital computation. Circulation,
Vol. 55, pp. 329-337, ISSN 0009-7322
Camici, P.; Prasad, S. & Rimoldi, O. (2008). Stunning, hibernation, and assessment of
myocardial viability. Circulation, Vol. 117, pp. 103-114, ISSN 1524-4539
Chipkin, S.; Frid, D.; Alpert, J.; Baker, S.; Dalen, J. & Aronin, N. (1987). Frequency of painless
myocardial ischemia during exercise tolerance testing in patients with and without
diabetes mellitus. Am J Cardiol, Vol. 59, pp. 61-55, ISSN 0002-9149
Das, M.; Aronow, W.; McClung, J. & Belkin, R. (2006). Increased prevalence of coronary
artery disease, silent myocardial ischemia, complex ventricular arrhythmias, atrial
fibrillation, left ventricular hypertrophy, mitral annular calcium, and aortic valve
calcium in patients with chronic renal insufficiency. Cardiol Rev, Vol. 14, pp. 14-17,
ISSN 1061-5377
Davies, M. (2000). The pathophysiology of acute coronary syndromes. Heart, Vol. 83, pp.
361-366, ISSN 1355-6037
Dodge, J.; Brown, B.; Bolson, E. & Dodge, H. (1988). Intrathoracic spatial location of specified
coronary segments on the normal human heart. Applications in quantitative
arteriography, assessment of regional risk and contraction, and anatomic display.
Circulation, Vol. 78, pp. 1167-1180, ISSN 0009-7322
Dodge, J., Brown, B.; Bolson, E. & Dodge, H. (1992). Lumen diameter of normal human
coronary arteries. Influence of age, sex, anatomic variation, and left ventricular
hypertrophy or dilation. Circulation, Vol. 86, pp. 232-246, ISSN 0009-7322
Felker , G.; Shaw, L. & O'Connor, C. (2002). A standardized definition of ischemic
cardiomyopathy for use in clinical research. J Am Coll Cardiol, Vol. 39 pp. 210-218,
ISSN 0735-1097
Friesinger, G.; Page, E. & Ross, R. (1970). Prognostic significance of coronary arteriography.
Trans Assoc Am Physicians, Vol. 83, pp. 78-92. ISSN 0066-9458
Funabashi, N.; Kobayashi, Y.; Perlroth, M. & Rubin, G. (2003). Coronary artery: quantitative
evaluation of normal diameter determined with electron-beam CT compared with
cine coronary angiography initial experience. Radiology, Vol. 226, pp. 263-271, ISSN
0033-8419
Garg, S.; Sarno, G.; Garcia-Garcia, H.; Girasis, C.; Wykrzykowska, J.; Dawkins, K.; et al.
(2010). A new tool for the risk stratification of patients with complex coronary
artery disease: the Clinical SYNTAX Score. Circ Cardiovasc Interv, Vol. 3, pp. 317-
326, ISSN 1941-7632
Gensini, G.; Kelly, A.; Da Costa, B. & Huntington, P. (1971). Quantitative angiography: the
measurement of coronary vasomobility in the intact animal and man. Chest, Vol. 60,
pp. 522-530, ISSN 0002-9149
Gensini G. (1983). A more meaningful scoring system for determining the severity of
coronary heart disease. Am J Cardiol, Vol. 51, p. 606, ISSN 0012-3692
Ghali, J.; Liao, Y.; Simmons, B.; Castaner, A.; Cao, G. & Cooper, R. (1992). The prognostic
role of left ventricular hypertrophy in patients with or without coronary artery
disease. Ann Intern Med, Vol. 117, pp. 831-836, ISSN 0003-4819
Gibson, C.; Cannon, C.; Murphy, S.; Marble, S.; Barron, H. & Braunwald, E. (2002).
Relationship of the TIMI myocardial perfusion grades, flow grades, frame count,
and percutaneous coronary intervention to long-term outcomes after thrombolytic
administration in acute myocardial infarction. Circulation, Vol. 105, pp. 1909-1913,
ISSN 1524-4539 (Electronic)
Summarized Coronary Artery Caliber and Left Ventricle
Mass for Scoring of Cardiac Ischemia: Diagnostic and Prognostic Value 295

Gibson, C.; Goel, M.; Murphy, S.; Dotani, I.; Marble, S.; Deckelbaum, L. et al. (2000). Global
impairment of coronary blood flow in the setting of acute coronary syndromes (a
RESTORE substudy). Randomized efficacy Study of Tirofiban for Outcomes and
Restenosis. Am J Cardiol, Vol. 86, pp. 1375-1377, A5, ISSN 0002-9149
Gibson, C.; Murphy, S.; Rizzo, M.; Ryan, K.; Marble, S.; McCabe, C.; et al. (1999).
Relationship between TIMI frame count and clinical outcomes after thrombolytic
administration. Thrombolysis in Myocardial Infarction (TIMI) Study Group.
Circulation, Vol. 99, pp. 1945-1950, ISSN 1524-4539 (Electronic)
Goldstein, J.; Demetriou, D.; Grines, C.; Pica, M.; Shoukfeh, M. & O'Neill, W. (2000).
Multiple complex coronary plaques in patients with acute myocardial infarction. N
Engl J Med, Vol. 343, pp. 915-922, ISSN 0028-4793
Graham, M.; Chambers, R. & Davies, R. (1999). Angiographic quantification of diffuse
coronary artery disease: reliability and prognostic value for bypass operations. J
Thorac Cardiovasc Surg, Vol. 118, pp. 618-627, ISSN 0022-5223
Hamsten, A.; Walldius, G.; Szamosi, A.; Dahlen, G. & de Faire, U. (1986). Relationship of
angiographically defined coronary artery disease to serum lipoproteins and
apolipoproteins in young survivors of myocardial infarction. Circulation, Vol. 73,
pp. 1097-1110, ISSN 0009-7322
Jalal A. ( 2007). An objective method for grading of distal disease in the grafted coronary
arteries. Interact Cardiovasc Thorac Surg, Vol. 6, pp. 451-455, ISSN 1569-9285
(Electronic)
Jenkins, P.; Harper, R. & Nestel, P. (1978). Severity of coronary atherosclerosis related to
lipoprotein concentration. Br Med J, Vol. 2, pp. 388-391, ISSN 0007-1447
Kalbfleisch, S.; McGillem, M.; Pinto, I.; Kavanaugh, K.; DeBoe, S. & Mancini, G. (1990).
Comparison of automated quantitative coronary angiography with caliper
measurements of percent diameter stenosis. Am J Cardiol, Vol. 65, pp. 1181-1184,
ISSN 0002-9149
Koren, M.; Devereux, R.; Casale, P.; Savage, D. & Laragh, J. (1991). Relation of left
ventricular mass and geometry to morbidity and mortality in uncomplicated
essential hypertension. Ann Intern Med, Vol. 114, pp. 345-352, ISSN 0003-4819
Korosoglou, G.; Haars, A.; Michael, G.; Erbacher, M.; Hardt , S.; Giannitsis, E. et al. (2007).
Quantitative evaluation of myocardial blush to assess tissue level reperfusion in
patients with acute ST-elevation myocardial infarction: incremental prognostic
value compared with visual assessment. Am Heart J, Vol. 153, pp. 612-620, ISSN
1097-6744 (Electronic)
Krone, R.; Laskey, W.; Johnson, C.; Kimmel, S.; Klein, L.; Weiner, B. et al. (2000). A
simplified lesion classification for predicting success and complications of coronary
angioplasty. Registry Committee of the Society for Cardiac Angiography and
Intervention. Am J Cardiol, Vol. 85, pp. 1179-1184, ISSN 0002-9149
Kucher, N.; Lipp, E.; Schwerzmann, M.; Zimmerli, M.; Allemann Y. & Seiler, C. (2001).
Gender differences in coronary artery size per 100 g of left ventricular mass in a
population without cardiac disease. Swiss Med Wkly, Vol. 131, pp. 610-615, ISSN
1424-7860
Kunadian, V.; Harrigan, C.; Zorkun, C.; Palmer, A.; Ogando, K.; Biller, L. et al. (2009). Use of
the TIMI frame count in the assessment of coronary artery blood flow and
microvascular function over the past 15 years. J Thromb Thrombolysis, Vol. 27, pp.
316-328, ISSN 1573-742X (Electronic)
296 Advances in the Diagnosis of Coronary Atherosclerosis

Kwon, S.; Kim, Y.; Shim, J.; Sung, J.; Han, M.; Kang, D. et al. (2011). Coronary artery calcium
scoring does not add prognostic value to standard 64-section CT angiography
protocol in low-risk patients suspected of having coronary artery disease. Radiology,
Vol. 259, pp. 92-99, ISSN 1527-1315 (Electronic)
Leaman, D.; Brower, R.; Meester, G.; Serruys, P.; van den Brand M. (1981). Coronary artery
atherosclerosis: severity of the disease, severity of angina pectoris and
compromised left ventricular function. Circulation, Vol. 63, pp. 285-299, ISSN 0009-
7322
Marcus, M.; Koyanagi, S.; Harrison, D.; Doty, D.; Hiratzka, L. & Eastham, C. (1983).
Abnormalities in the coronary circulation that occur as a consequence of cardiac
hypertrophy. Am J Med, Vol. 75, pp. 62-66, ISSN 0002-9343
Min, J.; Lin, F.; Dunning, A.; Delago, A.; Egan, J.; Shaw, L. et al. (2010). Incremental
prognostic significance of left ventricular dysfunction to coronary artery disease
detection by 64-detector row coronary computed tomographic angiography for the
prediction of all-cause mortality: results from a two-centre study of 5330 patients.
Eur Heart J, Vol. 31, pp. 1212-1219, ISSN 1522-9645 (Electronic)
Molloi, S.; Kassab, G. & Zhou, Y. (2001). Quantification of coronary artery lumen volume by
digital angiography: in vivo validation. Circulation, Vol. 104, pp. 2351-2357, ISSN
1522-9645 (Electronic)
Ringqvist, I.; Fisher, L.; Mock, M.; Davis, K.; Wedel, H.; Chaitman, B. et al. (1983). Prognostic
value of angiographic indices of coronary artery disease from the Coronary Artery
Surgery Study (CASS). J Clin Invest, Vol. 71, pp. 1854-1866, ISSN 0021-9738
(Linking)
Roberts, C. & Roberts, W. (1980). Cross-sectional area of the proximal portions of the three
major epicardial coronary arteries in 98 necropsy patients with different coronary
events. Relationship to heart weight, age and sex. Circulation, Vol. 62, pp. 953-959,
ISSN 0009-7322
Seiler, C.; Kirkeeide, R. & Gould, K. (1992). Basic structure-function relations of the
epicardial coronary vascular tree. Basis of quantitative coronary arteriography for
diffuse coronary artery disease. Circulation, Vol. 85, pp. 1987-2003, ISSN 0009-7322
Selwyn, AP. (1990). Current technology in assessing painless and painful ischemia. Am Heart
J, Vol. 120, pp. 722-725, ISSN 0002-8703
Sianos, G.; Morel, M.; Kappetein, A.; Morice, M.; Colombo, A.; Dawkins, K. et al. (2005). The
SYNTAX Score: an angiographic tool grading the complexity of coronary artery
disease. EuroIntervention, Vol. 1, pp. 219-227, ISSN 1774-024X
Sullivan, D.; Marwick, T. & Freedman, S. (1990). A new method of scoring coronary
angiograms to reflect extent of coronary atherosclerosis and improve correlation
with major risk factors. Am Heart J, Vol. 119, pp. 1262-1267, ISSN 0002-8703
Vogt, M.; Motz, W. & Strauer, B. (1992). Coronary haemodynamics in hypertensive heart
disease. Eur Heart J, Vol. 13 Suppl D, pp. 44-49, ISSN 0195-668X
Zaacks, S.; Allen, J.; Calvin, J.; Schaer, G.; Palvas, B.; Parrillo, J. et al. (1998 ). Value of the
American College of Cardiology/American Heart Association stenosis morphology
classification for coronary interventions in the late 1990s. Am J Cardiol, Vol. 82, pp.
3-9, ISSN 0002-9149
Zipes, D. &Wellens, H. (1998). Sudden cardiac death. Circulation, Vol. 98, pp. 2334-2351,
ISSN 0009-7322
16

Woven Coronary Artery


Ayşe Yıldırım1 and A. Deniz Oğuz2
1Kosuyolu Heart and
Research Hospital,
Pediatric Cardiology, Istanbul
2Gazi University Medical Faculty

Pediatric Cardiology, Ankara


Turkey

1. Introduction
In this chapter we will discuss the woven coronary artery which is one of the most
interesting coronary arterial anomalies. Although it is difficult to estimate the real incidence
of coronary artery anomalies due to the difficulty in specific identification, it is
approximately %0.3 - %1.3. The main reasons of underestimation of coronary artery
anomalies may be that, in many individuals; don’t lead to symptoms, morbidity, or
mortality (Friedman & Silverman, 2010).

2. Woven coronary artery


Woven coronary artery is an extremely rare congenital malformation characterized by the
division of epicardial coronary artery into thin channels which then reanastomose with the
distal part of the abnormal coronary artery. Generally this anomalous segment of the
coronary artery is limited to several centimeters long. By means of several angiographic
studies it has been shown that the blood flow is completely normal in all segments of the
woven coronary artery. This is a totally benign malformation which doesn’t lead to any
pathologic cardiac event.
Woven coronary artery was first described by Sane et al. in 1988, during the coronary
angiography performed on a 55 year old woman with congestive heart failure seven years
after from her aortic valve replacement and mitral valve commissurotomy (Sane, 1986). He
defined this branching and reanastomosing structure as ‘’figure 8’’pattern and named this
congenital anomaly as ‘’woven coronary artery’’.
Approximately nine adult and only one pediatric patient with woven coronary artery
have been reported since then. Most of these patients were males diagnosed accidentally
at the coronary angiography performed due to chest pain, after acute myocardial
infarction or due to the suspicion of left coronary artery aneurysm in Kawasaki disease.
Although the pathology is mostly seen in the right coronary artery, woven structures have
also been demonstrated in left coronary arteries or in circumflex artery. These are listed in
Table 1.
298 Advances in the Diagnosis of Coronary Atherosclerosis

AVR: aortic valve replacement, MVC: mitral valve commissurotomy, CHF: Congestive heart failure,
LAD: left anterior descending artery, RCA: right coronary artery, LCA: left coronary artery, AMI: acute
miyocardial infarction, LCXA: Left circumflex artery
Table 1. The summary of the demographic and clinical data of the patients with woven
coronary artery in the literature.
Woven Coronary Artery 299

2.1 Embryology of coronary artery


As there are very few number of woven coronary artery cases, the embryological
development of this rare coronary artery anomaly has not been explained yet. However,
there are two models of explanation for the embryologic development of normal coronary
arteries. Previously it had been thought that the cells of the myocardium nourished from the
blood in the ventricular cavity thicken and become close to the ventricular cavity by means
of multiple trabeculations. These trabeculations then develop into sinusoids which were the
forerunners of the coronary vascular system (Tomanek et al, 1996). The new model of
coronary arterial development suggests that the cells of the primordial liver form a
proepicardial protrusion. These cells establish the proepicardium and the epicardial cells
then migrate over the surface of the heart. The epicardial cells invade the subepicardial
matrix and form the coronary vascular plexus. By an undefined mechanism, probably
involving multiple growth factors, the epicardial cells undergo epithelial mesenchymal
transformation and form mature vessels. These small vessels on the surface of the heart fuse
and grow inward to penetrate the aorta fusing with the coronary vessels. The new
experimental data on the development of the coronary system suggests that beside multiple
growth factors, several adhesion molecules and chemotactic factors play role in this
complicated coordinated migration and transformation of cells to form coronary vessels.
The presence of congenital anomalies of coronary arteries may be due to abnormalities in
these signaling pathways or alterations in local factors that direct coronary vessel
development. (Matherne & Lim, 2008)

2.2 Etiology of woven coronary artery


The number of case studies published about woven coronary artery is so limited that the
etiology of this rarely encountered anomaly has not been clearly understood yet. The
reported cases on this anomaly are mostly adult patients. Recently a case report about an
infant nine- months-old with woven coronary artery has been published (Yıldırım et al.,
2010). Therefore it is now suspected that woven coronary artery may be a congenital
malformation and advanced studies are needed to lighten this issue

2.3 Incidence of woven coronary artery


The real incidence of woven CA is not known. The diagnosis of the patients in the literature
have been made accidentally so far. A remarkably high number of woven CA cases might
have been misdiagnosed as coronary artery thrombus, stenosis or dissection. It can be
claimed that the frequency of this anomaly could have been higher if woven coronary artery
anomaly had not been misinterpreted in this way.

3. Clinical features of woven coronary artery


As mentioned above ‘’woven coronary artery’’ is accepted as a benign condition. Most of the
woven coronary artery patients in the literature are the cases diagnosed unexpectedly
during the coronary angiography made upon acute myocardial infarction or angina
pectoris. In the case report of İyisoy at al., (2010) severe stenosis in the middle segment of
left anterior descending coronary artery and multiple thin channels in the right coronary
artery were demonstrated during the same coronary angiography session, however stenosis
300 Advances in the Diagnosis of Coronary Atherosclerosis

was accused to be responsible from the myocardial ischemia. Another coronary


angiography performed on a patient with positive stress test by Kursaklıoğlu et al. (2006)
revealed stenosis in the mid-segment of the right coronary artery and the division of arterial
lumen into multiple thin channels in the mid-segment of the circumflex artery. They
thought that the severe stenosis in the right coronary artery was the most possible reason of
the positive stress test in this patient rather than the woven coronary anomaly. In the cases
reported by Martuscelli et al.( 2000), Kursaklıoglu et al.(2006) and Yıldırım et al.(2010) no
adverse coronary events occurred during 4 to 5 year follow-up periods. Repeated control
coronary angiograms of the patients with coronary arterial stenting or cardiac surgery have
revealed no structural change in woven coronary arteries. This finding suggests that the
cause of chest pain, angina pectoris, acute myocardial infarction or positive stress test is not
dependent on woven coronary artery anomaly.

4. Diagnosis of woven coronary artery


Although there is an anatomically abnormal coronary artery, the blood flow and the
myocardial contraction is normal. Therefore the physical examination, laboratory findings,
chest x-rays, electrocardiographic and echocardiographic investigations of the woven
coronary artery patients are completely normal. The echocardiography is not helpful for the
diagnosis. The gold standard for the diagnosis of woven coronary artery is coronary
angiography. There is no angiographic uniform image of woven coronary arteries however
the most important angiographic criteria is definite intraluminal globular filling defects in
repeated angiographic views.

4.1 Cardiac catheterization


In the coronary angiography it is seen that the arterial lumen is divided into multiple thin
channels at the proximal segment of affected coronary arteries or their branches;
subsequently, these channels proceed distally with a slightly twisting route and then fuse
again. Distal to this woven segment, the downstream blood flow is absolutely normal
(Figures 1A, B and C).
During the coronary angiography division of coronary artery may be misdiagnosed as a
complicated plaque with thrombus formation instead of coronary artery malformation. In
addition, coronary artery stenosis, spontaneous coronary artery dissection, intracoronary
thrombosis or recanalization of a thrombus may mimic this woven structure. These
mimicking pathologies have lead to the misdiagnosis of woven coronary artery (Martuscelli
et al., 2000; Kaya et al., 2006). However for an accurate explanation of the etiology, more
detailed information about the characteristics and the development of this malformation is
needed.

5. Differential diagnosis
In differential diagnosis of woven coronary artery the important criteria is the absence of
coronary artery disease history of the patient with normal stress tests such as treadmill,
echocardiography and myocardial scintigraphy. The patient with a woven coronary artery
anomaly usually demonstrates normal coronary reserve during stress test.
Woven Coronary Artery 301

Fig. 1A. and B. Right coronary angiography showed proximal thin channels and distal
reanastomosis
This is dependent on normal blood flow distal to the anomalous segment (Kursaklıoglu et
al., 2006; Yıldırım et al., 2010; İyisoy et al., 2010). In woven coronary artery anomaly, normal
blood flow can be maintained distal to the anomalous segment, therefore coronary blood
reserve is not disturbed with stress tests, especially myocardial perfusion imaging (İyisoy et
al., 2101). Even if there is clinical evidence of myocardial infarction or angina pectoris in a
patient with woven coronary artery, coronary angiography should be repeated to eliminate
other coronary artery pathologies. Radiologically, the examination and the interpretation of
the angiographic image are crucial to prevent misdiagnosis of the woven coronary arteries.
In this coronary anomaly, although the filling is defective, flow is normal; which
differentiates this structure from other pathologies such as intracoronary thrombus, stenosis
or dissection.
302 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 1C. Diagram of the right woven coronary artery showing proximal thin channels (TC)
and distal reanastomosis (DRA)

6. Associated diseases
Up to now, woven coronary artery cases have only been reported as isolated coronary artery
anomaly (Sane et al., 1988; Berman et al.1990; Gregorini et al., 1995; Martuscelli et al., 2000;
Kursaklıoglu et al., 2006; Kaya et al., 2006; İyisoy et al., 2010). Only in the case report of
Yıldırım et al., (2010) the woven coronary artery was described as the part of a systemic
disease, Kawasaki disease, in an infant patient of nine months old. In this patient, routine
echocardiographic evaluation of coronary arteries revealed aneurysm of the left coronary
artery, leading to an angiographic study in which a woven right coronary artery was
detected coincidentally. In fact there is also a tendency for the development of thrombus in
Kawasaki disease and this condition should not be confused with woven coronary artery
(Yıldırım et al., 2010). For this reason, the pediatric and adult cardiologists should be aware
that woven coronary artery may accompany Kawasaki Disease and they should perform
coronary angiography to distinguish real thrombus from this coronary anomaly.

7. Treatment
Woven coronary artery is an anatomic abnormality and it doesn’t disturb cardiac functions
at all. Therefore it is not necessary to treat this anomaly. However other coronary artery
Woven Coronary Artery 303

pathologies such as coronary artery stenosis, thrombosis, dissection or myocardial infarction


should be carefully distinguished from woven coronary artery. This point is crucial to
prevent misdiagnosis and treatment of woven coronary artery unnecessarily.

8. Conclusion
Angiographic examination of the patients with suspected coronary artery pathologies
should be performed carefully to make differential diagnosis of dissection, thrombosis
stenosis and woven coronary artery. The catheter angiography scenes should be evaluated
repeatedly not to misinterpret the coronary artery pathologies. Such a detailed evaluation
and definitive diagnosis is important to reduce the risk of unnecessary percutaneous
coronary interventions, which expose the patients to several complications. Woven coronary
artery does not need treatment but follow-up is required as there are only a few reports
about the progress of this rare event. Advanced studies are needed on this unknown entity
to demonstrate its detailed pathology. Even though this anomaly appears to be a benign
coronary anomaly without any major adverse cardiovascular events, we need more data to
figure out its exact natural history.

9. Acknowledgment
As the current collected data about the woven coronary arteries has been provided from a
very limited number of cases, we believe that the embryology, pathophysiology, clinical
feature and the prognosis of this rare coronary anomaly will be explained better with
advanced studies.

10. References
Berman AD, Kim D, Baim DS.(1990) ’’Woven’’ right coronary artery: case report and
therapeutic implications. Cathet Cardiovasc Diagn ; 21:258–259.
Gregorini L, Perondi R, Pomidossi G, Saino A, Bossi IM, Zanchetti A. (1995). Woven left
coronary artery disease. Am J Cardiol ;75(4): 311-2.
Friedman AH and Silverman NH. Congenital anomalies of the coronary artery. (2010)
Pediatric Cardiology In Anderson RH, Baker EJ, Penny D, Redington, AN, Rigby ML,
Wernovsky G. (third edition), Churchill Livingstone and Elsevier, Philadelphia
pp:933-943
Iyisoy A, Celik T, Yuksel UC, Isik E. Woven right coronary artery: a case report and review
of the literature. Clin Cardiol. 2010;33:43-45.
Kaya D, Kilit C, Onrat E. (2006)An uncommon congenital anomaly of coronary arteries
misdiagnosed as intracoronary thrombus: woven coronary artery disease. Anadolu
Kardiol Derg. 6:383-384.
Kursaklioglu H, Iyisoy A, Celik T. (2006) Woven coronary artery: a case report and review of
literature. Int J Cardiol 113(1) 121-3.
Martuscelli E, Romeo F, Giovannini M, Nigri A. (2000) Woven coronary artery:
differential diagnosis with diffuse intracoronary thrombosis. Ital Heart J.
1(4):306-7.
304 Advances in the Diagnosis of Coronary Atherosclerosis

Matherne GP and Lim DS. Congenital anomalies of the coronary vessels and the aortic
root (2008). Moss and Adams’ Heart Disease in Infant, Children and
Adolescents including the Fetus and Young Adult. In Allen HD, Driscoll DJ,
Shaddy Re, Feltes TE. (seventh edition) Lippincott Williams & Wilkins,
Philadelphia. Pp:702-715.
Sane DC, Vidaillet JH Jr. (1988) Woven right coronary artery: a previously undescribed
congenital anomaly. Am J Cardiol 61(13):1158.
Tomanek RJ.(1996) Formation of the coronary vasculature: A brief review. Cardiovasc Res 31
Spec No: E46-51
Tomanek RJ. (2005) Formation of the coronary vasculature during development.
Angiogenesis 8:273-284
Yıldırım A, Oğuz D, Olguntürk R (2010). Woven Right and Aneurysmatic Left Coronary
Artery Associated with Kawasaki Disease in a 9-month-old Patient. Cardiol Young
20:342-344.
17

Image Post-Processing and Interpretation


Masahiro Jinzaki, Minoru Yamada and Sachio Kuribayashi
Dept. of Diagnostic Radiology, Keio University School of Medicine
Japan

1. Introduction
Image post-processing is defined as the process of integrating a series of axial images into a
form that is often easier to interpret than the sections themselves. The source transverse section
is a fundamental image for interpretation. However, because of the complexity of coronary
anatomy, interpreters must review coronary CT angiography interactively on workstations
capable of 3-dimensional (3D) displays. The 3D data set can be reformatted according to the
needs of the investigator using several types of post-processing algorithms. Coronary artery
interpretation includes assessment of calcium scoring on non-contrast image and coronary
angiography on enhanced image. Non-coronary cardiovascular findings and extra-cardiac
findings should be reviewed in addition to recognize primary and secondary comorbid
pathology and to identify findings that lead to alternative non-cardiovascular diagnoses. The
ultimate objective of post-processing and interpretation is to convey diagnostic information to
the treating physician with as much clarity and accuracy as possible. Interpretation times
vary, usually depending on the image quality and the user’s experience and knowledge of
cardiac CT and the individual workstation. Normally, experienced users usually spend no
more than 10 minutes segmenting and interpreting cardiac CT studies.

2. Post-processing technique
The source transverse sections are the primary tools for interpretation of coronary CT
angiography examinations. Post-processing imaging is performed from reconstructed axial
images and is useful in allowing clinically relevant information to be extracted from large
number of axial images. In addition, the diagnostic accuracy of axial images (86% sensitive
and 78% accurate) for the detection of coronary stenosis is low compared with combination
of post-processing techniques (98% sensitive and 94% accurate) (Jinzaki et al, 2009). Thus,
the review of 3D displays is necessary for the coronary artery interpretation due to the
complexity of coronary anatomy. These include volume rendering, multiplanar reformation,
thin-slab maximum intensity projection, curved multiplanar reformation, angiographic
view, and plaque-loaded angiographic view. A post-processing image which enables the
evaluation of coronary artery disease with fewer images and one that is understandable to
the third person is preferable.

2.1 Multiplanar reformation (MPR)


MPR is a high-resolution reconstruction format that allows display of planar images at any
angular section through the acquisition volume, which permits visualization in not only the
306 Advances in the Diagnosis of Coronary Atherosclerosis

axial plane but also in orthogonal (coronal and sagittal) or oblique planes that better follow
the arterial course in the thorax. The slice thickness can be modified, and a thick slab may be
helpful in visualizing tortuous coronary artery. MPR is a simple algorithm which can be
rendered quickly and accurately on any image-processing workstations. The distance
measurements in MPR are accurate. Different structures in MPR are accurate and do not
overlap. There is no loss of voxel value information due to threshold.
MPRs are best suited for evaluation of the individual cardiac chambers, aorta (Fig. 1), and
pulmonary vasculature with cardiac CT. MPRs are also well suited for selective evaluation
of a coronary short segment. If a narrowing is suspected on axial views, display in coronal
or sagittal orientations can confirm the presence of a stenosis (Fig. 2).
The disadvantage of MPR is that only short segments can be analyzed at a time due to the
nonlinear pathway of the coronary arteries. Thus, MPR requires the creation of numerous
images in different planes at every suspected location of coronary stenosis. Furthermore, it
is less suited for displaying the length of an artery. Grading of a stenosis is not advisable
with coronal, sagittal, or oblique reconstructions alone, because of partial averaging
(Johnson et al, 2010).

Fig. 1. Multiplanar reformation of cardiac structure


Oblique coronal image orientate better the planes of the geometrical orientations of cardiac
structure (left chamber and aorta).

a) b) c)
Fig. 2. Multiplanar reconstruction of coronary stenosis
Image Post-Processing and Interpretation 307

When a narrowing (arrow) is suspected on axial views (a), multiplanar reformation can be
used to set the planes (b, c) in an orientation that matches one of the coronary short
segments.

2.2 Thin-slab maximum intensity projection (Thin-slab MIP)


In both thin-slab MIP and MPR, orthogonal or oblique planes can be reviewed interactively.
MIP is a projection technique, in which only the highest density pixel in the projection
direction is displayed to reconstruct the two-dimensional images. MIP image allows the
differentiation between enhanced vascular structures and non-vascular structures, because
the result is a projection of the highest density. In practice, MIP is usually performed
interactively with a sliding slab, enabling the radiologist to adjust the window, select the
optimal orientation to display each artery, and modify the slab thickness (Fishman et al,
2006).
The advantage of thin-slab MIP is the creation of a vascular map quickly without
modification of several parameters (Fig. 3). In addition, MIP is better for displaying small-
caliber segments: the minimum vessel diameter seen with MIP was 0.7 mm versus 1.5 mm
for volume rendering (VR) (Zhang et al, 2004). This is because MIP is designed to selectively
demonstrate the arterial vasculature, which involves selection of the highest attenuation
voxels along rays projected through the volume. It is controversial whether thin-slab MIP
can be used for the diagnosis of coronary artery stenosis. One study compared
interpretation of 3 sets of orthogonally oriented thin-slab MIPs with axial sections with 16-
slice CT (Choi et al, 2006). For all three radiologists, the Az (area under the ROC curve)
values from the MIP images were higher than those Az values from the transaxial images,
however, the difference was significant for only one reader.
MIP has a tendency to overestimate stenosis. Heavily calcified arteries cannot be evaluated
with MIP. Thus, thin-slab MIP cannot be used for the quantity evaluation of plaque,
including the determination of grading of a stenosis. In addition, numerous images in
different planes are required for the evaluation of the whole coronary arteries. Thin-slab
MIP is useful as an adjunct to VR. However, it should not be the sole technique used for
interpretation.

a) b) c)
Fig. 3. Thin-slab maximum intensity projection of coronary stenosis
When a narrowing (arrow) is suspected on axial views (a), the use of thin-slab maximum
intensity projection (b) increases the overview of each coronary artery. Thin-slab maximum
intensity projection enables creation of a vascular map (c) quickly without modification of
several parameters.
308 Advances in the Diagnosis of Coronary Atherosclerosis

2.3 Curved multiplanar reformation (curved MPR)


The development of curved MPR format made it possible for the interpreter to follow the
course of a tortuous vessel for longer distances as it changes direction (Fig. 4) (Ferencik et
al., 2007). The artery can be displayed as a straight line (Fig. 5). This method enables one
entire coronary artery to be followed and displayed in one reconstruction, and reduces the
number of planar images needed, compared with MPR. An automated program is possible
to track the coronary arteries using placement of a single point in any portion of each artery
or ascending aorta, the computer can trace a given coronary vessel departing proximally
and distally from the point chosen, and extract the images in seconds. Other automated
program tracks the path of the coronary artery, when the operator selects two points: one at
the beginning and one at the end of the vessel. These applications also provide MPRs that
are orthogonal to the vessel centerline, thus creating cross-sectional cuts of each coronary
arterial segment (Fig. 5). All other surrounding structures are automatically eliminated that
are any distance from this centerpoint, possibly decreasing problems seen with other
reconstruction methods from tortuosity and overlap.

a) b)
Fig. 4. Curved multiplanar reformation of the coronary arteries.
The vessel can be seen in a long length with this technique. Fig. 4a demonstrates a
multiplanar reformation of the entire right coronary artery, including a segment of
posterolateral marginal branch. The atrio ventricular node artery (arrow) would require a
second reconstruction to follow that artery. The left anterior descending (b), with multiple
diagonal branches, often needs multiple reconstructions to demonstrate its entire course.
Curved MPR with coronary CT angiography has been reported with high sensitivity and
specificity (Achenbach et al., 1998). Significant stenosis and occlusions of the coronary
arteries can be visualized with an accuracy that exceeds 90%, since the CT value is
preserved, making it fairly easy to delineate calcifications from the lumen. Recognition of
calcifications with either higher or lower density than inner lumen contrast is very difficult
with VR, somewhat better with MIP, and best with MPR. In addition, curved MPR is used to
identify and quantify the degree of stenosis without being hindered by partial averaging
(Fig. 5). This method is also useful in delineating the morphology of plaque and its effect on
the lumen and adjacent vessel wall. Furthermore, the distance measurements in the curved
MPR images represent the true distance. Overall, curved MPR, because they can be
rendered quickly and convey all the information about the different CT densities within the
vessel, is an extremely useful tool to evaluate contrast-enhanced CT angiography data.
Image Post-Processing and Interpretation 309

The drawback of curved MPR is that only one vessel can be visualized in one image and that
side branches are not depicted unless separate reconstructions are rendered for every side
branch (Achenbach et al., 1998). Angiographic view is better in evaluating multiple
branches. Furthermore, accurate assessment for stenosis requires correct centerline
placement. Since the quality and ability of the computer workstation for tracking the vessel
depends on border definition and overall image quality, off-axis centerline placement may
happen, which result in misinterpretation of stenosis. Thus, curved MPR requires some
editing in most cases. This commonly occurs in the presence of heavy arterial calcification
and at the site of origin of branch.

a)

b) c) d)
Fig. 5. Curved multiplanar reformation for coronary artery stenosis
Coronary stenosis (arrow) is demonstrated in the proximal segment of the right coronary
artery. The observer can always create a plane orthogonal to the direction of the vessel,
which allows the evaluation of the configuration of plaque and grading of a stenosis. The
three orthogonal planes are at the level of a proximal normal lumen (b), a significant
stenosis (c), and a distal normal lumen (d), respectively.

2.4 Volume rendering (VR)


Rendering is a term taken from computer graphics and represents the process of generating
a 3D model from a 2D image. Opacity and color are assigned to each CT value interval via
transfer function, which is selectable and interactively modifiable (Calhoun et al., 1999). For
example, normal soft tissue is assigned high transparency, enhanced vascular structures
slight opaqueness and bone strong opaqueness. The sum of all CT values along each search
ray from the observer through the 3D data volume, weighted by the transfer function, is
displayed. In addition, color is used in most cases, with the color intensity decreasing with
increasing distance to the observer in order to generate depth information and a 3D
impression. The image can be rotated to different views, allowing the interpreter some
flexibility in seeing the segment from multiple angles (Fig. 6).
310 Advances in the Diagnosis of Coronary Atherosclerosis

VR provides an excellent overview of the cardiac and vascular anatomy, such as defining
the course of coronary anomalies (Fig. 7a), the presence and course of coronary bypass
grafts (Fig. 7b), the analysis of thoracic cardiovascular anatomy and congenital heart disease
(Fig. 7c). Because VR creates volumetric 3D representations with the illusion of spatial
integrity, it is helpful for surgeons to understand the anatomic complexity before surgery,
and for teaching purposes and as illustrations for patients.
It is controversial whether VR can be used for the assessment of coronary stenosis. One
study using 16 slice CT compared 3D VRs with combination of techniques including axial
images, MPRs, and curved MPRs and MIPs. Sensitivity (63% vs 74%), specificity (80% vs
76%), and accuracy (78% vs 75%) were not significantly different between VR and the
combination of techniques, respectively (Cordeiro et al., 2006). However, the diameter of
vessels and the apparent thickness of the vessel lumen are dependent on threshold or
transparency settings and the computer algorithm that is used to subtract non-vascular
structures with VR (Ferencik et al., 2007) (Fig. 8). Furthermore, the overlap of cardiac veins
on coronary arteries often hinders the coronary artery stenosis at the overlapped segment on
VR (Fig. 9). Another disadvantage of VR is that due to its use of threshold or transparency
setting which obscure the lower attenuation structures, the visualization of smaller vessel
may be inferior compared with other displaying methods (Johnson et al, 2010). VR also
requires several angles to evaluate the entire coronary artery (Fig. 6).

a) b)

c) d)
Fig. 6. Volume rendering of the coronary arteries.
Volume rendering provides an excellent overview of coronary anomalies, the course of
coronary bypass grafts, and congenital heart disease
a. Coronary anomalies: both coronary arteries originate from left coronary cusp (stent is
placed in the proximal segment of left descending artery).
b. Bypass grafts: right internal thoracic artery (RITA) graft communicate with the left
descending artery and left internal thoracic artery (LITA) graft communicate with
obtuse marginal branch.
Image Post-Processing and Interpretation 311

c. Major aortopulmonary collateral artery in patients with Tetralogy of Fallot (red: aortic
arch, yellow: right pulmonary artery arising from aorta, green: left pulmonary arteries
arising from ductus arteriosus).

a) b) c)
Fig. 7. Volume rendering of cardiac and vascular anatomy
Volume rendering allows an easy overview of coronary artery anatomy. It assists
significantly in understanding the topography of coronary vessels and position of the lesion
in coronary tree. Dedicated projections for each of the three main vessels are performed. The
proximal and middle segment of the RCA is visualized in a view from the right side (a). The
distal segment of the RCA is visualized in a right caudal view (b). The left anterior
descending artery is visualized in a left anterior view (c). The left circumflex artery is
visualized in a left cranial view and a left view (d).

a) b) c)
Fig. 8. The affect of modifying threshold and opacity setting on volume rendering
Coronary stenosis (arrow) is suspected in the proximal segment of left descending artery in
the image with high opacity setting (a), while it is difficult to detect coronary stenosis in the
image with low opacity setting (b). Significant coronary stenosis (c: arrow) is seen in curved
MPR at the same location suspected in volume rendering with high opacity setting. The
diameter of coronary artery is also modified by changing the threshold and opacity setting.
312 Advances in the Diagnosis of Coronary Atherosclerosis

a) b) c)
Fig. 9. Overlapped coronary vein on volume rendering
Coronary stenosis in the proximal left circumflex artery (arrow) is hindered by overlapped
great cardiac vein on volume rendering (a), while it is well visualized on angiographic view
(b: arrow) and curved MPR (c: arrow).

2.5 Angiographic view (AGV)


AGV is an whole heart MIP image in which contrast media of the ventricles is eliminated
(Fig.10). This image is similar to that from invasive conventional coronary angiography (CAG).
In our institute, AGV images divided into right and left coronary artery were rendered
interactively from at least three angles for right coronary artery and six angles for left coronary
artery (Fig. 11). If necessary, reconstructions were rendered from additional angles to visualize
the entire coronary tree. AGV was originally automatically created only in GE workstation, but
is available in many other workstations. It can be created in 3 steps after VR of cardiac CT is
created in the usual method (Jinzaki et al., 2006). First, the origins of the right and left coronary
arteries are separated from the ascending aorta. Second, the image of the ascending aorta and
left ventricle is selected and singled out by clicking any area of the ascending aorta. Third, this
image is subtracted from the original cardiac image, which leaves only the image of the
coronary artery. Similar images can be created by tracking and extracting the coronary artery
itself; however, tracking or extracting requires the threshold of CT attenuation, so the images
can vary depending on the threshold and the ability of the workstation being used. AGV keeps
the coronary artery untouched, and thus AGV does not vary among workstations.

a) b) c)
Fig. 10. Angiographic view
Image Post-Processing and Interpretation 313

Angiographic view image clearly demonstrates the distribution of coronary artery stenosis
(long arrow) and coronary calcifications (short arrows) in one image. This resembles the
images of coronary angiography and is understandable by third parties. Angiographic view
image (a) and coronary angiography (b, c) shows considerable consistency in the detection
of coronary artery stenosis (long arrows).

a) b) c)

d) e) f)

g) h) i)
Fig. 11. Angiographic view divided into right and left coronary artery
Angiographic view image divided into right and left coronary artery enables viewing the
coronary arteries with the same angle as CAG. We create three directions in RCA [a: left
anterior oblique (LAO) 45 and cranial angulations (CRA) 0, b: LAO 20 and CRA 30, c: right
anterior oblique (RAO) 30 and CRA 0) and 6 directions in LCA (d: LAO 0 and CRA 30, e:
LAO 45 and CRA 25, f: RAO 20 and CRA 40, g: RAO 30 and caudal angulations (CAU) 30, h:
RAO 0 and CAU 30, i: LAO 50 and CAU 30].
314 Advances in the Diagnosis of Coronary Atherosclerosis

The advantage of AGV image is that it clearly demonstrates the distribution of high-density
lesions, such as coronary calcifications and stents, and coronary artery stenosis in a single
image, making it easier to be understood when viewed by any third party. Because the
structures are displayed based upon their brightness, calcium and metal are easily
distinguished from the contrast-enhanced lumen. For the detection of coronary stenosis,
AGV was equally sensitive (98%) and accurate (91%) to a combination of conventional
techniques, including interactive MIP and MPR as well as preset curved MPRs
(98%sensitive, 94% accurate) (Jinzaki et al., 2009). AGV is also better for displaying small-
caliber segments as described in thin-slab MIP. Furthermore, AGV divided into right and
left coronary artery enables viewing the lesion with the same angle as CAG, and the
detection of best angle of the lesion prior to PCI (Fig. 12). Collateral arteries are also well
visualized in AGV. The whole collateral arteries are demonstrated with AGV in less images
than volume rendering, since AGV is a projection image (Fig. 13). Thus, AGV would be
useful for explaining the severity of disease to the patient, as a guide for the reading due to
its high diagnostic accuracy, and in the discussion of the treatment strategy in conference.

Fig. 12. The detection of best angle of the lesion prior to PCI
The coronary stenosis (arrow) is best visualized in the view of RAO 30 and CAU 30
(middle). This information will help cardiologists in discussing treatment strategy and
reduce the number of CAG image acquisitions.

a) b)
Fig. 13. Angiographic view in case with collateral vessels
In patients with chronic total occlusion of RCA, collateral vessels are seen between left
descending artery and posterior descending artery (yellow arrows), left circumflex artery
and posterolateral artery (blue arrows). These collateral vessels are well demonstrated in
one image with angiographic view (a), while the whole collateral arteries are not visualized
in one image with volume rendering (b).
Image Post-Processing and Interpretation 315

A disadvantage of AGV is that since it is a 2D display, it requires review of multiple rotated


images to understand 3D relationships. MIP does not provide in-depth information or
attenuation detail within the slice, because overlapped higher-density structures obscure the
lower-density structures of interest. Static MIP images can be misleading, by incorrect
placement of arteries. Another disadvantage is that MIP algorithm is prone to artifacts. One
well-known pitfall is the ‘‘string of beads’’ appearance, because a normal small vessel
passing obliquely through a volume is only partially represented by voxels along its length
(Calhoun et al., 1999). Because MIP selects the highest density voxels along a ray projected
through the volume, adequate contrast enhancement is imperative. MIP has a tendency to
overestimate the stenosis, especially in case with calcification (Rubin et al., 1994). Non-
calcified plaque may be overlooked unless it creates significant luminal narrowing, because
of its low attenuation value.

2.6 Plaque-loaded angiographic view (PLAG)


So far, there is no post-processing technique that can display an overview image of the
lumen and vessel wall of a whole heart in one image (Fig. 14). PLAG is the image in which
the non-calcified plaque image is merged with the AGV (Jinzaki et al., 2008). As a result,
PLAG allows the noninvasive visualization of the whole coronary lumen and the vessel wall
in one image. PLAG is now available in GE workstation and some other workstations. If this
is not available in your workstation, it can be generated as follows: First, the color-coded
plaque image of each coronary artery is created (Fig 14a). The CT attenuation of the
coronary lumen is usually larger than 250HU, while that of non-calcified plaque is typically
less than 120HU. Setting the color code range at less than 120 HU can generate a “plaque
image”, in which only the vessel wall and the non-calcified plaques are color-coded. Second,
AGV image of the entire coronary artery (Fig 14b), which is similar to that shown by a CAG
is separately generated, which displays the distribution of calcified plaques as well as the
lumen in more detail. Third, the color-coded “plaque image” in curved MPR is re-formatted
into a MIP image to merge with AGV image. Finally, loading the MIP “plaque image” onto
AGV image completes the process (Figs 14c). The critical factors in PLAG are that the
merged images are both in MIP format, and taking advantage of a relatively large CT
attenuation discrepancy between the lumen and non-calcified plaque allows color-coding,
which helps to better visualize plaques when loaded onto AGV.
PLAG will provide comprehensive information for coronary CT angiography in discussing
the treatment strategy of both coronary stenosis and non-stenotic non-calcified plaques. This
image also enables the overview of the tortuous vessel in the chronic total occlusion (CTO),
which would be useful for the percutaneous revascularization of CTO (Fig.15).
In future, with the further improvement of temporal and spatial resolution of MDCT, and
with enhanced accuracy of plaque volume quantification is enhanced, this image could also
be useful in evaluating and demonstrating the therapeutic effect of anti-atherosclerotic
drugs in patients with multiple soft plaques in coronary arteries (the so-called vulnerable
patients) on one image.
The characteristics of each post-processing
For the effective use of the various post-processing techniques, knowledge of the
characteristics of each post-processing method is mandatory. The strengths and weaknesses
of the available post-processing technique are summarized in Table 1.
316 Advances in the Diagnosis of Coronary Atherosclerosis

a) b) c) d)
Fig. 14. Plaque-loaded Angiographic View of multiple non-calcified plaques
Curved MPR of the right coronary artery (a) demonstrates the non-stenotic non-calcified
plaques (arrows), which are assigned blue in color-coded image (b). The angiographic view
image (c) demonstrates the lumen of the entire coronary artery. The color-coded image in
curved MPR is re-formatted into a MIP image and merged with the Angiographic View
image. The Plaque-loaded Angiographic View image (d) demonstrates the lumen and
multiple plaques (arrows) of coronary arteries in one image.

a) b)
Fig. 15. Plaque-loaded Angiographic View of chronic totally occlusion
An angiographic view image (a) could not provide the information of the occluded lesion.
The Plaque-loaded Angiographic View image (b) demonstrates the overview of the tortuous
vessel (arrows) in the occluded lesion.

3. Image interpretation
At the beginning of interpretation, an overview of the data set should be performed to
detect artifacts, gross abnormalities and the presence of non-coronary cardiac or extra-
cardiac abnormalities. Coronary artery interpretation is then performed using post-
processing images
Image Post-Processing and Interpretation 317

Table 1. Comparison of post-processing techniques

3.1 Examination of image quality


It is imperative that an interpreter spend the first minute making sure the dataset is
adequate. Artifacts due to motion, calcification, and metallic densities; image noise; and
poor contrast enhancement may degrade the quality of the study as well as simulate or
obscure coronary stenoses (Achenbach et al., 2003, Ferencik et al, 2003). To evaluate the
image quality, the axial CT source images, multiplanar reformatted images, and also three-
dimensional images were analyzed. The reader should specifically state if an artery or artery
segment is not interpretable and why.
318 Advances in the Diagnosis of Coronary Atherosclerosis

3.1.1 Artifact
“Artifact” can be defined as any discrepancy between the reconstructed Hounsfield values
in the image and the true attenuation coefficients of the object in such a way that these
discrepancies are clinically significant or relevant as judged by the radiologist (Hsieh, J.,
2003). Artifacts of coronary CT angiography were classified into three categories (Table 2).
The first is cardiac motion-related artifact (Choi et al., 2004). This is caused by many factors,
including a heart rate greater than 70–75 beats per minute during imaging acquisition,
variations in heart rate during breath holding (Fig 16), arrhythmia, and inappropriate
selection of pitch or reconstruction window. Artifacts are classified into two patterns:
motion blurring or stepladder effects. Blurring occurs when movement in the cardiac
structure of interest exceeds the temporal resolution of scanning, either because of a fast
heart rate or because of an inappropriate selection of the reconstruction window for the
particular coronary artery. Stepladder artifacts are due to motion occurring between
reconstruction of sequential heartbeats. The easiest way of reducing cardiac motion artifacts
is to lower the heart rate by the prior administration of-blocker. Customized reconstructions
at a different cardiac phase may be successful by either adjusting the phase of reconstruction
or removing data from undesirable beats.

a) b)
Fig. 16. Stepladder artifact due to changing heart rate during CT acquisition.
Volume-rendered image (a) shows a severe motion-related artifact (arrows) caused by
acceleration in heart rate toward the end of the breath hold. Selection of an optimal
reconstruction window reduced the artifact (b).
The second is voluntary motion that is generally preventable with careful instruction of the
patient (Choi et al., 2004). Artifacts due to breathing (Fig 17) or body motion are distinctive
because they affect the bones of the anterior or lateral chest wall in addition to the coronary
arteries; these are less likely to be correctable by additional reconstructions. Oxygen
supplementation may help dyspneic patients hold their breaths for a longer period.
The third is high-attenuating artifact (Choi et al., 2004). High-attenuating artifact includes,
blooming artifacts, beam hardening artifacts and streaking artifacts. Blooming artifact is a
result of partial-volume effects, causing the status that small high-contrast objects appear
larger than they are. These artifacts commonly make calcified plaque (Fig 18a) and stents
appear to narrow the lumen more than they actually do. Use of appropriate reconstruction
filters may reduce these artifacts. Beam hardening presents as dark banding between dense
objects. These are also seen in calcified plaque and stents (Fig 18b). Streaking artifact is a
kind of reconstruction artifact due to undersampling, photon starvation, motion, beam
Image Post-Processing and Interpretation 319

hardening, or scatter. In coronary CT angiography, these are usually caused by metallic


objects such as clips, markers, and wires used in coronary artery bypass surgery, and often
called as metal artifact (Fig 18c).

a) b)
Fig. 17. Stepladder artifact due to breathing
Sagittal maximum intensity projection image (a) and coronal MPR image (b) show a
severe respiration-related artifact in the anterior chest wall (arrow) and at the diaphragm
(arrow).

a) b) c)
Fig. 18. High-attenuating artifacts
a. Blooming artifact caused by severe coronary calcifications that interfere with the
evaluation of luminal stenosis.
b. Beam hardening artifacts caused by coronary stent that prevents accurate evaluation of
the stent lumen.
c. Metal artifacts caused by surgical clips.

3.1.2 Image noise


Improper scan parameter (low tube output for a given body size) causes poor signal-to-
noise (Fig 19). This is mainly due to obesity. Improper reconstruction during a part of the
cardiac cycle with reduced tube current from EKG-guided tube modulation also results in
low image quality.
320 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 19. Low image quality due to improper scan parameters.


Poor signal-to-noise is caused by low tube output for a given body size

3.1.3 Low contrast enhancement


The methods of reconstruction rely on Hounsfield units (HU) to make images. Values of 250
HU and above are ideal, and current protocols generally yield mean enhancement levels in
the range of 300 to 350 HU for right and left coronary arteries (Johnson et al., 2009, Husmann
et al., 2009). Low contrast attenuation may be secondary to improper image acquisition
timing or slow contrast injection.

Table 2. Various artifacts in coronary CT angiography


Image Post-Processing and Interpretation 321

3.2 Non-coronary cardiac findings


Non-coronary cardiovascular structures include the pericardium, cardiac chambers,
interatrial septum, interventricular septum, atrioventricular valves, ventriculo-arterial
valves, pulmonary arteries, pulmonary veins, thoracic aorta, imaged aortic branch arteries,
and central systemic veins. The axial images or MPRs is useful for a general understanding
of the spatial relationship of these structures and abnormality. Left ventricular and left atrial
myocardial walls and chamber cavities should be examined for hypertrophy, dilation,
thinning (Fig. 20a), hypodense enhancement, thrombus (Fig 20b), masses, and congenital
anomalies. These structures should be reviewed within the cardiac field-of-view and any
abnormalities described.

a) b)
Fig. 20. Non-coronary cardiac findings
a. Focal thinning of myocardial wall due to ventricular diverticula (arrow)
b. Thrombus in ventricular chamber (arrow)

3.3 Extra-cardiac structures


Extra-cardiac structures within the field of view of routine coronary CT angiography
include the mediastinum, hilum, trachea and bronchi, lung parenchyma, pleura, chest wall,
esophagus, stomach, liver, spleen, and colon. Interpretation of the axial images or MPR
provides a general understanding of the anatomic relationships of the heart and coronary
arteries with surrounding extra-cardiac structures. Review of all visible non-cardiovascular
structures is important for two principal reasons: (1) recognition of primary and secondary
comorbid pathology and (2) identification of findings that lead to alternative non-
cardiovascular diagnoses.

3.4 Coronary artery interpretation


In analyzing coronary arteries, a systematic approach using axial images and post-processing
images should be followed. Realizing from the outset that no single method of analysis is
without limitations, the incorporative review of the 2D images and the different 3D renderings
is necessary. However, how to use the various available post-processing techniques remains a
matter of personal experience and may differ among various operators.
322 Advances in the Diagnosis of Coronary Atherosclerosis

3.4.1 Interpretation of axial images


The axial image evaluation is a fundamental and essential step, because information from
the original data set (axial images) can be lost as a consequence of processing the images for
advanced postprocessing methods. Scrolling through the axial images in the cranio-caudal
direction allows interpretation of the anatomy and gross abnormality of coronary arteries.
Myocardial bridge of coronary artery is often easily noticed in axial images than post-
processing images (Fig 21a). However, following each segment on each level (over 50-
80contiguous images) may be difficult, and the evaluation of coronary stenosis is often more
difficult compared with the post-processing methods (Jinzaki et al., 2009) (Fig 21b). For
example, correct diagnosis of coronary stenosis is difficult with the axial images in the
lesions located in the segment running horizontal to the axial section. Diffuse concentric
plaque with positive remodeling also has a tendency to be incorrectly viewed as stenotic on
axial images, possibly because the reference diameter (vessel diameter in non-diseased
artery immediately proximal to the lesion) of these lesions is not measurable on the axial
image. The post-processing image is better suited for evaluation of these types of lesions.

a) b)
Fig. 21. Interpretation of axial images
Myocardial bridge of coronary artery (a: arrow) is often easily noticed in axial images. The
diagnosis of coronary stenosis is easy in the lesions located in the segment running the
cranio-caudal direction (b: arrow), however difficult in the lesions located in the segment
running horizontal to the axial section.

3.4.2 The use of post-processing images


After the interpretation of axial images, the readers should shift to post-processing images.
VR provide an overview of the coronary tree. Also, more complex anatomy (such as
congenital heart disease or bypass grafts) is easily seen. For coronary artery evaluation, the
combination of several types of post-processing algorithms such as thin-slab MIP, MPR and
curved MPR is usually used. However, these methods require numerous images to evaluate
the whole coronary artery, while AGV would be easy to overview the distribution of
coronary lesion. Thus, as an initial guide for the interpretation, we recommend first to use
AGV. Furthermore, in the comparison of the AGV image and combination of several types
of post-processing, sensitivity was the same (98%) and there was no significant difference in
accuracy (Jinzaki et al., 2009). If coronary artery stenosis is detected on AGV, the lesion
Image Post-Processing and Interpretation 323

should be evaluated further for vessel wall morphology and composition using curved
MPR. Further evaluation could be omitted in branch vessels with no stenotic lesions on
AGV, since the negative predictive value of AGV is very high (99%) and evaluating multiple
diagonal branches or obtuse marginal branches is time-consuming with curved MPR or
other methods. However, even if stenotic lesion is not detected in three main arteries on
AGV, we recommend reconstructing curved MPR of these arteries to evaluate non-stenotic
non-calcified plaque, which may be missed on AGV. This is because acute myocardial
infarction is frequently related to a segment that was not significantly stenosed at previous
angiography (Littele, et al., 1988, Giroud et al., 1992). Acute coronary occlusions leading to
myocardial infarction tend to cluster in predictable "hot spots" within the proximal third of
the coronary arteries (Wang et al., 2004). Identification of low attenuation plaques in these
high-risk zones for acute coronary occlusions will be important for potentially locally
directed preventive strategies.

Fig. 22. Workflow of post-processing and interpretation for coronary artery evaluation
Step 1. Overview of coronary artery on angiographic view
Step 2. Quantitative evaluation of coronary lesions detected on angiographic view
Step 3. Evaluation of three main arteries using curved MPR and no further evaluation in
branch vessels without stenotic lesions on angiographic view
324 Advances in the Diagnosis of Coronary Atherosclerosis

3.4.3 The guiding principles of interpretation


To analyze coronary arteries a systemic approach should be followed. It is recommended
that the coronary segments are evaluated in a sequential order from segment 1 to segment
15 following the standard report of the American Heart Association. The interpretation of
coronary arteries include (1) assessment of stenosis severity, (2) vessel wall morphology and
composition, (3) total occlusions, and (4) bypass graft and stent.
a. Assessment of stenosis severity
The description of luminal stenosis grading and plaque length is important in the
interpretation. The degree of coronary stenosis is manually calculated by subtracting vessel
diameter at the site of maximal stenosis from the mean of the vessel reference segment
diameters (proximal and distal references) and then dividing the difference by the mean of
the vessel reference segment diameters on curved MPR and cross-sectional image (Fig 22).
Recent cardiac- vessel analysis packages enables calculating percent stenosis automatically
when the user selects the area of maximum narrowing and normal vessel proximal and
distal to the stenosis (Busch et al., 2007). However, current spatial resolution of CT has not
demonstrated sufficient reproducibility or accuracy in predicting findings of CAG to make
such measurements a routine requirement. Thus, at the present time, Society of
Cardiovascular Computed Tomography guideline recommends that arterial segments be
described within broad stenosis ranges (Cheng et al., 2008, Raff et al., 2009) (Table 3),
although future technical developments may improve the precision of stenosis
quantification.

Table 3. Quantitative Stenosis Grading


b. Vessel wall morphology and composition
Vessel wall morphology and composition are important information of coronary CT
angiography, since these are not available on CAG. coronary CT angiography can
visualize intramural presence of positively remodeled plaque and differentiate calcific,
non-calcific, and mixed plaque. Society of Cardiovascular Computed Tomography
recommends that the description of plaques as ‘‘non-calcific’’ is preferable to ‘‘soft’’ or
‘‘lipid-rich’’ since low CT density (in Hounsfield units) levels do not necessarily correlate
closely with anatomic pathology or biochemistry (Raff et al., 2009). Recent studies
revealed that the CT characteristics of plaques associated with ACS include positive
vascular remodeling, low plaque density<30HU, and spotty calcification (Motoyama et
al., 2009) (Fig. 23). Thus, the description of these features will assist interpretation. The
Image Post-Processing and Interpretation 325

vascular remodeling index is defined as lesion diameter/reference diameter, and is


reported as positive when the diameter at the plaque site was at least 10% larger than the
reference segment. Spotty calcification was defined when the calcification is <3 mm in
size. Features of plaque morphology such as ulceration, dissection, and fissuring are also
noted when image quality is sufficient.

a) b) c)
Fig. 23. Low attenuation plaque and spotty calcification
Curved MPR demonstrates significant stenosis in left descending artery (a: arrow). The
lesion is accompanied with spotty calcification (b: arrow) and area < 30HU (c: arrow).

a) b) c)
Fig. 24. Positive remodeling
Non-calcified plaque (a: long arrows) in right coronary artery is positively remodeled as
compared with the normal coronary segment proximal to the lesion (short arrows). The
diameter was 4.3 mm in normal segment (b) and 4.9 mm in the lesion (c). Remodeling index
was 1.14.
326 Advances in the Diagnosis of Coronary Atherosclerosis

c. Total occlusions
PCI of CTO has a lower success rate than PCI of non-occluded coronary stenosis. Failure of
guide-wire crossing the occluded lesion is the main cause of unsuccessful PCI of CTO.
Coronary CT angiography may provide useful information for selecting patients suitable for
PCI of CTO (Soon et al., 2007). It can show the collateral supply in the distal vessel beyond
the occlusion (Fig. 13). The proximal and distal ends of an occluded segment can be
distinctively seen on coronary CT angiography. Therefore, the occlusion length can be
measured on CCTA (Fig. 24). Mollet et al. also reported that CTO lesion length > 15 mm
measured on CT was associated with higher failure rate of PCI (Mollet et al., 2005). Heavy
calcification of CTO has been reported to be an independent predictor of failed PCI of CTO
(Noguchi et al., 2000). The exact location of calcified plaque on the image of coronary CT
angiography may help the interventionist to steer the guide wire away from the calcified
lesion that is not well seen on conventional angiography. PLAG images are able to
demonstrate the overview of occluded lesion, which are helpful in providing the roadmap
for interventionists during the process of wiring the occluded vessel (Fig. 15).

a) b)
Fig. 25. CT evaluation of chronic total occlusion
The total occlusion is seen in the distal segment of right coronary artery on thin-slab MIP
images (a: arrows). The occlusion length can be measured on stretched MPR (b). Calcified
plaque is also well visualized.
Image Post-Processing and Interpretation 327

d. Bypass grafts, stents


The evaluation of coronary bypass grafts by coronary CT angiography is highly accurate
in predicting the findings on invasive coronary angiography (Meyer et al., 2007). The
location and anastomoses of bypass grafts should be described in addition to the location
and severity of stenoses. The evaluation of stents by MDCT is often difficult due to
blooming artifact and beam hardening effects. The evaluation of in-stent stenosis depends
highly on stent size and composition. Stents 3.0 mm or larger (85%) were judged
assessable; 26% of stents smaller than 3 mm were judged assessable (Schroeder et al.,
2008). Different stent compositions and structures appeared to be associated with
variations in the extent of residual artifact. The use of special reconstruction algorithms
improved visualization of the stent lumen and reduced blooming artifact with a drawback
of a modest increase in noise in the images (Maintz et al., 2006).

4. Conclusion
Many post-processing images are available for the evaluation of coronary artery. The
interpreter should know the advantage and disadvantage of each post-processing
image, and perform the incorporative review of the 2D images and the different 3D
renderings.

5. References
Achenbach, S., Moshage, W., Ropers, D., & Bachmann, K. (1998). Curved multiplanar
reconstructions for the evaluation of contrast-enhanced electron beam CT of the
coronary arteries. AJR Am J Roentgenol, Vol.170, No.4, (Apr 1998), pp. 895-899.
Achenbach, S., Giesler, T., Ropers, D., Ulzheimer, S., Anders, K., Wenkel, E., Pohle, K.,
Kachelriess, M., Derlien, H., Kalender, WA., Daniel, WG., Bautz, W., & Baum, U.
(2003). Comparison of image quality in contrast-enhanced coronary-artery
visualization by electron beam tomography and retrospectively electrocardiogram-
gated multislice spiral computed tomography. Invest Radiol, Vol.38, No.2, (Feb
2003), pp. 119–128.
Busch, S., Johnson, TR., Nikolaou, K., von Ziegler, F., Knez, A., Reiser, MF., & Becker, CR.
(2007). Visual and automatic grading of coronary artery stenoses with 64-slice CT
angiography in reference to invasive angiography. Eur Radiol, Vol.17, No. 6, (Jun
2007), pp.1445-1451.
Cademartiri, F; Mollet, NR & Nieman, K. (2005). Image post-processing, In Computed
Tomography of the Coronary Arteries, Feyter PJ & Krestin GP, (ed.), 27-46, Taylor &
Francis, New York
Calhoun, PS., Kuszyk, BS., Heath, DG., Carley, JC., & Fishman, EK. (1999). Three-
dimensional volume rendering of spiral CT data: theory and method. Radiographics,
Vol.19, No.3, (May-Jun 1999), pp. 745-764.
Cheng, V., Gutstein, A., Wolak, A., Suzuki, Y., Dey, D., Gransar, H., Thomson, LEJ., Hayes,
SW., Friedman, JD., & Berman, DS. (2008). Moving beyond binary grading of
coronary arterial stenoses on coronary computed tomographic angiography:
insights for the imager and referring clinician. J Am Coll Cardiol Imaging, Vol.1, No.
4, (Jul 2008), pp. 460-471.
328 Advances in the Diagnosis of Coronary Atherosclerosis

Choi, HS., Choi, BW., Choe, KO., Cho,i D., Yoo, KJ., Kim, MI., & Kim, J. (2004). Pitfalls,
artifacts, and remedies in multi- detector row CT coronary angiography.
Radiographics, Vol.24, No.3, (May-Jun 2004), pp. 787-800.
Choi, JW., Seo, JB., Do, KH., Choi, SI., Lee, W., Ko, SM., Lee, SH., Lee, JS., Song, JW., Song,
KS., & Lim, TH. (2006). Comparison of transaxial source images and 3-plane, thin-
slab maximal intensity projection images for the diagnosis of coronary artery
stenosis with using ECG-gated cardiac CT. Korean J Radio,l Vol.7, No.1, (Jan-Mar
2006), pp. 20-27.
Cordeiro, MA., Lardo, AC., Brito, MS., Rosário Neto, MA., Siqueira, MH., Parga, JR., Avila,
LF., Ramires, JA., Lima, JA., & Rochitte, CE. (2006). CT angiography in highly
calcified arteries: 2D manual vs. modified automated 3D approach to identify
coronary stenoses. Int J Cardiovasc Imaging, Vol.22, No.3-4, (Jun-Aug 2006), pp.507-
516.
Ferencik, M., Moselewski, F., Ropers, D., Hoffmann, U., Baum, U., Anders, K., Pomerantsev,
EV., Abbara, S., Brady, TJ., & Achenbach, S.(2003). Quantitative parameters of
image quality in multidetector spiral computed tomographic coronary imaging
with submillimeter collimation. Am J Cardiol, Vol.92, No.11, (Dec 2003), pp. 1257–
1262.
Ferencik, M., Ropers, D., Abbara, S., Cury, RC., Hoffmann, U., Nieman, K., Brady, TJ.,
Moselewski, F., Daniel, WG., & Achenbach, S. (2007). Diagnostic accuracy of image
postprocessing methods for the detection of coronary artery stenoses by using
multidetector CT. Radiology, Vol.243, No.3, (Jun 2007), pp. 696–702.
Fishman, EK., Ney, DR., Heath, DG., Corl, FM., Horton, KM., & Johnson, PT. (2006).
Volume rendering versus maximum intensity projection in CT angiography:
what works best, when, and why. Radiographics, Vol.26, No.3, (May-Jun 2006),
pp. 905-922.
Giroud, D., Li, JM., Urban, P., Meier, B., Rutishauer, W. (1992). Relation of the site of acute
myocardial infarction to the most severe coronary arterial stenosis at prior
angiography. Am J Cardiol, Vol.69, No.8, (Mar 1992), pp. 729-732.
Hsieh, J. (2003). Image artifacts: appearances, causes, and corrections. In Computed
tomography: principles, design, artifacts, and recent advance, Hsieh, J, (Ed.), 167–240,
SPIE Press, Bellingham, USA
Husmann, L., Gaemperl,i O., Valenta, I., Schepis, T., Scheffel, H., Stolzmann, P., Leschka, S.,
Desbiolles, L., Marincek, B., Alkadhi, H., & Kaufmann, PA. (2009). Impact of vessel
attenuation on quantitative coronary angiography with 64-slice CT. Br J Radil,
Vol.82, No.980, (Aug 2009), pp. 649-653.
Jinzaki, M., Sato, K., Tanami, Y., Yamada, M., Kuribayashi, S., Anzai, T., Asakura, Y., &
Ogawa, S. (2006). Novel method of displaying coronary CT angiography -
Angiographic view-. Circ J, Vol.70, No.12, (Dec 2006), pp. 1661-662.
Jinzaki, M., Sato, K., Tanami, Y., Yamada, M., Anzai, T., Kawamura, A., Ueno, K., &
Kuribayashi, S. (2009). Diagnostic accuracy of angiographic view image for the
detection of coronary artery stenoses by 64-detector row CT: a pilot study
comparison with conventional post-processing methods and axial images alone.
Circ J, Vol.73, No.4, (Apr 2009), pp. 691-698.
Jinzaki, M., Yamada, M., Sato, K., Tanami, Y., Anzai, T., Sasaki, K., & Kuribayashi, S.
(2008). Overview Image of the Lumen and Vessel Wall in Coronary CT
Image Post-Processing and Interpretation 329

Angiography -The Plaque-Loaded Angiographic View-. Circ J, Vol.72, No.4,


(Apr 2008), pp. 671-673.
Johnson, PT., Pannu, HK., & Fishman, EK. (2009). IV contrast infusion for coronary artery
CT angiography: literature review and results of a nationwide survey. AJR Am J
Roentgenol Vol.192, No.5, (May 2009), pp.W214–221.
Johnson, PT., & Fishman, EK. (2010). Postprocessing techniques for cardiac computed
tomographic angiography. Radiol Clin North Am, Vol.48, No.4, (Jul 2010), pp. 687-
700.
Little, WC., Constantinescu, M., Applegate, RJ., Kutcher, MA., Burrows, MT., Kahl, FR.,
Santamore, WP. (1988). Can coronary angiography predict the site of a subsequent
myocardial infarction in patients with mild-to-moderate coronary artery disease?
Circulation, Vol.78, No.5, (Nov 1988), pp. 1157-1166.
Maintz, D., Seifarth, H., Raupach, R., Flohr, T., Rink, M., Sommer, T., Ozgün, M.,
Heindel, W., & Fischbach, R.(2006). 64-slice multidetector coronary CT
angiography: in vitro evaluation of 68 different stents. Eur Radiol, Vol.16, No. 4,
(Apr 2006), pp. 818-826.
Meyer, TS., Martinoff, S., Hadamitzky, M., Will, A., Kastrati, A., Schomig, A., & Hausleiter,
J. (2007). Improved noninvasive assessment of coronary artery bypass grafts with
64-slice computed tomographic angiography in an unselected patient population. J
Am Coll Cardiol, Vol.49, No. 9, (Mar 2007), pp. 946-950.
Mollet, NR., Hoye, A., Lemos, PA., Cademartiri, F., Sianos, G., McFadden, EP., Krestin, GP.,
Serruys, PW., & de Feyter, PJ.(2005). Value of preprocedure multislice computed
tomographic coronary angiography to predict the outcome of percutaneous
recanalization of chronic total occlusions. Am J Cardiol, Vol.95, No. 2, (Jan 2005), pp.
240–243.
Motoyama, S., Sarai, M., Harigaya, H., Anno, H., Inoue, K., Hara, T., Naruse, H., Ishii, J.,
Hishida, H., Wong, ND., Virmani, R., Kondo, T., Ozaki, Y., & Narula, J. (2009).
Computed tomographic angiography characteristics of atherosclerotic plaques
subsequently resulting in acute coronary syndrome. J Am Coll Cardiol, Vol.54, No. 1,
(Jun 2009), pp. 49-57.
Noguchi, T., Miyazaki, MD S., Morii, I., Daikoku, S., Goto, Y., & Nonogi, H. (2000).
Percutaneous transluminal coronary angioplasty of chronic total occlusions.
Determinants of primary success and long-term clinical outcome, Catheter
Cardiovasc Interv Vol.49, No. 3, (Mar 2000), pp. 258–264.
Raff, GL., Abidov, A., Achenbach, S., Berman, DS., Boxt, LM., Budoff, MJ., Cheng, V.,
DeFrance, T., Hellinger, JC., Karlsberg, RP., & Society of Cardiovascular Computed
Tomography. (2006). SCCT guidelines for the interpretation and reporting of
coronary computed tomographic angiography. J Cardiovasc Comput Tomogr, Vol.3,
No. 2, (Mar-Apr 2006), pp.122-136.
Rubin, GD., Dake, MD., Napel, S., Jeffrey, RB Jr., McDonnell, CH., Sommer, FG.,
Wexler, L., & Williams, DM. (1994). Spiral CT of renal artery stenosis:
Comparison of three-dimensional rendering techniques. Radiology, Vol.190,
No.1, (Jan 1994), pp. 181–189.
Schroeder, S., Achenbach, S., Bengel, F., Burgstahler, C., Cademartiri, F., de Feyter, P.,
George, R., Kaufmann, P., Kopp, AF., Knuuti, J., Ropers, D., Schuijf, J., Tops,
LF., & Bax, JJ.; Working Group Nuclear Cardiology and Cardiac CT; European
330 Advances in the Diagnosis of Coronary Atherosclerosis

Society of Cardiology; European Council of Nuclear Cardiology. (2008).


Cardiac computed tomography: indications, applications, limitations, and
training requirements: report of a Writing Group deployed by the Working
Group Nuclear Cardiology and Cardiac CT of the European Society of
Cardiology and the European Council of Nuclear Cardiology. Eur Heart J,
Vol.29, No. 4, (Feb 2008), pp. 531-556.
Soon, KH., Selvanayagam, JB., Cox, N., Kelly, AM., Bell, KW., & Lim, YL. (2007).
Percutaneous revascularization of chronic total occlusions: review of the role of
invasive and non-invasive imaging modalities. Int J Cardiol, Vol.116, No. 1, (Mar
2007), pp. 1-6.
Wang, JC., Normand, SL., Mauri, L., & Kuntz, RE. (2004). Coronary artery spatial
distribution of acute myocardial infarction occlusions. Circulation, Vol.110, No.3,
(Jul 2004), pp. 278-284.
Zhang, ZH., Jin, ZY., Li, DJ., Lin, SB., Zhang, SY., Kong, LY., Wang, Y., Wang, LH.,
Zhao, WM., Mou, WB., Zhang, LR., Zhu, WL., Ni, C., Ren, H., Yu, HQ., Miao,
Q., & Fang, Q. (2004). Non-invasive imaging of coronary artery with 16-slice
spiral computed tomography. Chin Med Sci J, Vol.19, No.3, (Sep 2004), pp. 174-
179.
18

Novel Insights Into Stenosis on Coronary


Angiography–Outline of Functional
Assessment of Stable Angina
Patients with Angiographic Stenosis
Shinichiro Tanaka
The Second Department of Internal Medicine,
Gifu University Graduate School of Medicine, Gifu
The Department of Cardiology,
Gifu Prefectural General Medical Center,Gifu
Japan

1. Introduction
Coronary angiography is an invasive technique for imaging the coronary artery lumen and
remains one of the most accurate methods for diagnosing coronary artery disease.
Furthermore, this invasive technique is the standard method for guiding revascularization
procedures such as percutaneous coronary intervention (PCI) and coronary-artery bypass
graft (CABG), as well as for guiding stent placement during PCI. However, the degree of
coronary stenosis is usually decided by visual estimation of the percentage diameter
narrowing using the proximal assumed normal arterial as a reference. There is 20% variation
among readings of experienced angiographers, and the same angiographer may even render
a different interpretation at a time remote from the first reading. Borderline stenosis, or
angiographic narrowing of 40 to 70%, does not always correspond to abnormal physiology
and myocardial ischemia (1,2). For such lesions, noninvasive or direct physiological
measurements of impaired flow validate the decision to initiate revascularization therapy.
Though quantitative coronary angiography can be reproduced for assessment of coronary
stenosis, the inability to determine the functional severity of coronary stenosis remains a
limitation of coronary angiography.
Intracoronary physiological parameters have been introduced to assess functional coronary
lesion severity during cardiac catheterization. Measurement of coronary flow reserve (CFR;
coronary flow velocity response to adenosine) and fractional flow reserve (FFRmyo;
coronary pressure-derived with adenosine) provide information about functional severity
(3). Furthermore, FFRmyo, which is calculated from coronary pressure measurements, is an
invasive index used to identify a stenosis responsible for reversible ischemia (4,5,6).
For most patients with stable angina, the goal of treatment is complete, or nearly complete
elimination of anginal chest pain and return to normal activities with a functional capacity
corresponding to Canadian Cardiovascular Society (CCS) class I angina. This goal should be
accomplished with minimal side effects, as well as with a reduced risk of mortality (7). The
332 Advances in the Diagnosis of Coronary Atherosclerosis

topics of this chapter encompass the activities of daily life and functional stenosis in patients
with angiographically demonstrated stenosis.

2. Functional severity
For coronary angiographic assessment employing cardiac catheterization, it should be noted
that the stenotic lumen is compared to a nearby angiographically normal lumen, which may
still show diffuse atherosclerotic disease. Furthermore, lesions containing diffuse,
concentric, symmetrical disease, remodeling, or dissection, will be underestimated when
disease severity is assessed (Fig.1). CFR and FFRmyo, which provide information about
functional severity, can be performed during cardiac catheterization.

(A) Stenosis can be visualized from multiple angles. (B) Stenosis can be viewed as indicated by the
white arrow, but is normally visualized as indicated by the black arrow. Stricture necessitates changing
the angle. (C) Vascular conduits appear normal at all angles despite the existence of stenosis.
Fig. 1. Angiographic images and vascular conduits
CFR reflects both epicaudal and microvascular disease but does not identify these entities by
itself (8,9). Intra-coronary flow velocity can be measured with a 0.014-inch doppler guide
wire during baseline measurements and maximum hyperemia which is induced by
administering adenosine. CFR was calculated as the ratio of hyperemic to baseline of the
average peak velocity (10,11). CFR can also be measured with a temperature sensor-tipped
Novel Insights Into Stenosis on Coronary Angiography–Outline
of Functional Assessment of Stable Angina Patients with Angiographic Stenosis 333

guidewire, as the ratio of the inverse mean transit times obtained by thermodilution curves
at baseline and during hyperemia (12,13,14). The parameters of CFR show agreement at a
cut-off value of 2.0 (11,15).
On the other hand, FFRmyo is an accurate functional index of epicaudal stenosis (4,5). Intra-
coronary pressure can be measured with a 0.014-inch pressure-monitoring guidewire, which
was first calibrated, then set to be equal with the aortic pressure in the guiding catheter, and
finally positioned distal to the coronary lesion. Mean aortic and distal pressures were
obtained during baseline measurements, and FFRmyo was calculated as the ratio of the
mean distal pressure to the mean aortic pressure during maximum hyperemia (3,4,5).
Although a previous report emphasized the importance of combining pressure and flow
velocity measurements to evaluate coronary lesion severity and microvascular involvement
(11), FFRmyo is a reliable index of the functional severity of coronary stenosis, and an
FFRmyo value of 0.75 distinguishes stenoses associated with inducible ischemia from those
that are not. Indeed, in patients with stable chest pain, the most important prognostic factor
is the occurrence of myocardial ischemia reflected by an FFRmyo of < 0.75 (5,6, Fig.2).

In the case on the left, angiographic stenosis was 56% in the left circumflex branch (LCX) by quantitave
coronary angiography (A). During adenosine infusion, the wire was pulled back to track coronary
pressure from the far distal LCX to the catheter, and after crossing the lesion produced a change in
coronary pressure (FFRmyo = 0.58).
Likewise, in the case shown on the right, angiographic stenosis was 46% in left anterior descending
branch (LAD) by quantitative coronary angiography (C), and FFRmyo = 0.70 (D)
Fig. 2. Representative case of FFRmyo
334 Advances in the Diagnosis of Coronary Atherosclerosis

Even when treated with PCI, the clinical outcomes of these patients are significantly worse
than those of patients with functionally insignificant stenoses (FFRmyo  0.75) (16,17). When
FFRmyo was used to divide patients into a group in which stenosis was most likely to be
physiologically significant (FFRmyo < 0.75) and a group in which it was not, the overlap
between the 2 groups in terms of angiographic severity was so large that it suggested
angiography (16,17) could not be used to predict the absence or presence of inducible
ischemia in individual patients (4,18, Fig.3). In fact, however, in patients with multivessel
coronary artery disease undergoing PCI with drug eluting stents, routine measurement of
FFR in addition to angiographic guidance, as compared with PCI guided by angiography
alone, results in a significant reduction in major adverse events (19).

(A) The percent stenosis is likely to be physiological; FFRmyo. (B) The overlap between stenosis and
FFRmyo was large.

Fig. 3. Variations in percent stenosis on coronary angiography and FFRmyo

3. Functional capacity
The factors contributing to impaired exercise capacity appear to be multifactorial and
include increasing age, sedentary lifestyle, depression, obesity and other comorbidities as
well as impaired cardiovascular function. Poor exercise capacity is generally considered to
be a predictor of mortality in patients with ischemic heart disease as well as all cause
mortality (20,21). Traditional exercise tests such as the treadmill test or walking distance in 6
minutes are well-known methods for measurement of exercise capacity.
Additionally, self-assessed questionnaires, such as the Duke Activity Status INDEX (DASI)
(22), the Specific Activity Questionnaire (SAQ) (23) and the Veterans Specific Activity
Questionnaire (VSAQ) (24), can also measure exercise capacity as a well validated measure
of functional capacity that can be expressed as metabolic equivalents (METs) and have been
shown to correlate with adverse outcomes. Self-assessed questionnaires can be a simple and
easy tool that contributes to the global risk assessment (10).
The cardiopulmonary exercise test (CPX) is useful for measurement of excise capacity
employing various parameters at each stage in a series of exercises (Fig.4). A symptom-limited
incremental exercise test is usually performed using an upright, electromagnetically braked
cycle ergometer or treadmill with measurement of breath-by-breath VO2, carbon dioxide
Novel Insights Into Stenosis on Coronary Angiography–Outline
of Functional Assessment of Stable Angina Patients with Angiographic Stenosis 335

production (VCO2) and minute ventilation (VE). This evaluates peak VO2, oxygen uptake at
the anaerobic threshold (AT) and so on. A 12-lead electrocardiogram (ECG) and heart rate
were continuously monitored throughout the test, and cuff blood pressure was measured
every minute using an automatic manometer. CPX testing is more useful than self-assessed
health status, which is subjective and can vary among patients depending upon symptom
complexity and the unique perceptions, expectations and preferences of individuals (7).

Fig. 4. Representative case of CPX


336 Advances in the Diagnosis of Coronary Atherosclerosis

4. Functional severity and exercise capacity in patients with angiographic


stenosis
Coronary angiography employing cardiac catheterization has an important role in
revascularization procedures such as PCI, and PCI has become a common initial
management strategy for patients with stable angiographic stenosis. However, at present,
the initial management strategy of patients with stenosis on coronary angiography should
be to reduce not only the risks for major cardiac events but also to eliminate angina and
allow a return to normal activities. Previous studies have shown that PCI does not reduce
the long-term rates of death, myocardial infarction or other major cardiovascular events
with compared to optimal medical therapy alone (25). PCI can provide an incremental
benefit in quality of life compared with that provided by optimal medical therapy alone
among patients with chronic coronary artery disease for approximately 24 months (26).
Another previous study showed that in patients with stable coronary artery disease and an
angiographically documented stenosis treatable by PCI, a 12-month exercise-training
program resulted in a higher event-free survival rate and higher exercise capacity than
standard PCI (27). Additionally, PCI significantly increased exercise capacity values such as
AT and peak VO2 in patients with peak VO2 < 15 ml/kg/min, whereas it yielded no
significant improvement in those with peak VO2 ≥ 15 ml/kg/min (28). Based on these
observations, the existence of stenosis on coronary angiography is an insufficient indication
for revascularization. It has been reported that an increase in functional capacity as indicated
by self-assessed questionnaire is associated with an increase in CFR, although 86% of subjects
did not have significant stenosis on coronary angiography (10). Impaired overall functional
capacity is independently associated with coronary microvascular dysfunction or endothelial
dysfunction, among the most important determinants of myocardial ischemia. On the other
hand, routine FFRmyo measurement with angiographic guidance resulted in a significant
reduction in major adverse cardiac effects, even when patients underwent PCI with drug-
eluting stents (19). Furthermore, FFRmyo is associated with inducible ischemia which other
measures are not, thereby providing higher accuracy than for any other invasive or
noninvasive test. According to the aforementioned reports, it seemed that the existence of
stenosis on coronary angiography is a notwell validated measure of stable angina. FFRmyo
and functional capacity should be determined in stable angina patients with angiographic
stenosis. Previously, we assessed the relationship between FFRmyo and CPX values (4).
Our study subjects were 15 males (65.8+/-8.9 years old) with stable angina and 75%
angiographic stenosis in one coronary artery. Eligible patients had no evidence of acute
coronary syndromes, prior myocardial infarction, significant valvular heart disease, diabetes
mellitus treated with insulin, smoking; or occupational, orthopedic and other conditions
that precluded exercise. Neither did they have left main coronary artery stenosis > 25% or
high-grade proximal left anterior descending artery stenosis with significant stenosis (>
75%) of other vessels, nor left ventricular angiography-based detection of abnormal wall
motions such as asynergy or diffuse hypokinesis, or reduced left ventricular function
(ejection fraction < 55%). The %diameter stenosis (%DS) was determined to be 61.7+/-9.1%
based on quantitative coronary angiography. Mean FFRmyo was 0.84+/-0.66, peak VO2 was
17.1+/-3.2ml/kg/min and AT was 11.1+/-2.0ml/kg/min. There was no significant
correlation between %DS and FFRmyo (r = 0.12, p = ns, Fig.3A), peak VO2 (r = -0.051, p =
ns) or AT (r = -0.013, p = ns). By contrast, there was a significant positive correlation (Fig.5)
between FFRmyo and peak VO2 (r = 0.534, p < 0.05) and between FFRmyo and AT (r =
Novel Insights Into Stenosis on Coronary Angiography–Outline
of Functional Assessment of Stable Angina Patients with Angiographic Stenosis 337

0.542, p < 0.05). A previous study showed stable angina patients with impaired functional
capacity as assessed by a CPX test to likely have reduced FFRmyo. The present study also
showed that an FFRmyo value of 0.75 reflects stenoses associated with inducible ischemia
and hence appears to correspond to a peak VO2 value of 15.0. A cut-off value of FFRmyo of
0.75 nearly matched the exercise capacity value (peak VO2; 15 ml/kg/min) improvement
with PCI. As noted above, exercise capacity in patients with stable angina reflects functional
rather than angiographic stenosis. Exercise training can improve myocardial perfusion, as
revealed by myocardial scintigraphy, as well as maximal exercise capacity (27). Therefore,
that study suggested that it may be possible to improve FFRmyo by increasing exercise
capacity through training.

(A) There is no correlation between angiographic stenosis and peak VO2. However, there is a positive
correlation between FFRmyo and peak VO2 (B). (Source: Circ J 2009;73: 2308–2314)
Fig. 5. Relationship between exercise capacity and stenosis.

4.1 Case report


This is a representative case showing improvement in functional capacity despite the
existence of angiographic stenosis.
338 Advances in the Diagnosis of Coronary Atherosclerosis

A 58-year-old woman presented with unstable angina. Coronary angiography revealed 99%
stenosis of the right coronary artery (RCA#3), and 75% stenosis (#11) and 90% stenosis (#12)
of the left circumflex branch (LCX). She received stent implantation to #3 in the emergent
stage (Fig.6). Thallium-201 scintigraphy with adenosine revealed ischemia in the LCX area
(Fig.7A). PCI was recommended, but the patient refused because the condition was
asymptomatic in her daily life. This patient did not wish to undergo revascularization of the
LCX and continue to receive cardiac rehabilitation (physical training at AT level). Peak VO2
increased from13.3 ml/kg/min (Fig.4) to 16.8 ml/kg/min. Thallium-201 scintigraphy
(Fig.7B) with adenosine documented improved myocardial perfusion distal to the LCX.
(Ischemia area reduced from 13.7% to 8.75%)

There was (A) 99% stenosis of the right coronary artery (RCA #3) and a (B) stent was implanted. Left coronary
angiongraphy revealed 75% stenosis (#11) and 90% stenosis (#12) of the left circumflex branch (LCX).
Fig. 6. Coronary angiographies of case repor

Fig. 7. Bull’s eye image from thallium-201 single-photon emission computed tomography
(A) pre-training (ischemic area 13.7%), and (B) post-training (ischemic area 8.75%)
Novel Insights Into Stenosis on Coronary Angiography–Outline
of Functional Assessment of Stable Angina Patients with Angiographic Stenosis 339

5. Functional severity, exercise capacity and tissue characterization


Acute coronary syndrome is triggered by microscopic ulcerations of vulnerable
atherosclerotic plaques. The majority of vulnerable plaques appear “angiographically
insignificant” before their rupture (less than 75% diameter stenosis). In contrast, most of the
“significant” plaques (greater than 75% stenosis) visualized at angiography are at low risk
for plaque rupture (29).
Intravascular ultrasound (IVUS) allows cross-sectional imaging of coronary arteries and
provides a more comprehensive assessment of the atherosclerotic plaques in vivo (30, Fig 8).
Studies using IVUS have indicated that coronary atherosclerosis is underestimated when
visually analyzing angiographic results owing to coronary compensatory remodeling and
the diffuse nature of coronary atherosclerosis, which frequently makes the reference vessel
appear normal angiographically (31).

Intravascular ultrasound provides an image of an atheroma in a normal segment (B,E) as well as a


stenotic lesion (C,D) on angiography
Fig. 8. Representative angiographic and IVUS images
An integrated backscatter intravascular ultrasound (IB-IVUS) system has been developed in
which two-dimensional color-coded maps allowing plaque tissue characterization in
coronary arteries can be constructed by computer (32,33). A computer equipped with IB-
IVUS software is connected to the IVUS imaging system to obtain radio frequency signal
output, signal trigger output, and video image output. IB is calculated as the average power
340 Advances in the Diagnosis of Coronary Atherosclerosis

of the ultrasound backscattered signal from a small volume of tissue using fast Fourier
transform measured in decibels (dB) (32). IB-IVUS is a useful method for analyzing coronary
plaque tissue (Fig.9), which accurately and quantitatively differentiates among calcification,
dense fibrosis, fibrosis and lipid components (33). IB-IVUS results previously suggested that
classifying plaques as vulnerable or stable using this technique is an effective means of
predicting acute coronary syndrome. When evaluated using IB-IVUS, plaques are deemed
vulnerable when the lipid area is > 65% and the fibrotic area is < 25% (30). Thus, functional
stenosis and tissue characterization may be linked to clinical outcome. Previously, we
assessed the relationship between functional stenosis and the characterization of plaque
tissue using FFRmyo and IB-IVUS (18).

The initial image was constructed by tracing the lumen and the external elastic membrane. In the color-
coded map, red is calcification, yellow and green are fibrosis, and blue represents the lipid pool.
Fig. 9. Representative IB-IVUS images
We studied 17 lesions showing 75%-stenosis visually by coronary angiography in 17 stable
angina patients (64.2+/-9.1 years old, 11 males). We found no correlation between FFRmyo
and %DS (r = -0.354, p = 0.166, Fig.3B). Nor was there a correlation between FFRmyo and
plaque burden (r = -0.241, p = 0.359, Fig.1B), or between FFRmyo and minimum lumen
cross-sectional area (r = -0.002, p = 0.995). Likewise, neither %DS nor plaque burden
correlated with the tissue characterization values (Figs.10A). There was no correlation
between FFRmyo and %CA (calcification area) (r = -0.068, p = 0.799). By contrast, we
observed a significant positive correlation between FFRmyo and %F (r = 0.620 p = 0.0067),
and a significant negative correlation between FFRmyo and %LP (lipid pool area) (r = -0.524,
p = 0.0293) (Fig.10B).
Our previous findings indicate that the tissue characteristics of coronary plaques in
intermediate lesions affect functional stenosis. Fibrous tissue and FFRmyo also correlate
positively. Lipid pool and FFRmyo correlate negatively. The apparently contradictory
results obtained in this study might be explained by the progression of human
atherosclerotic lesions leading to increasing fibrosis and stenosis. Distal pressure,
additionally, decreases with the severity of coronary narrowing in the epicardial arteries,
Novel Insights Into Stenosis on Coronary Angiography–Outline
of Functional Assessment of Stable Angina Patients with Angiographic Stenosis 341

contributing to the progression of atherosclerosis. However, intermediate or moderate


lesions remain distinct in terms of functional and angiographic severity. The increase in
fibrous tissue is likely an adaptive response to this distal pressure aimed at redistributing
and modulating the mechanical stress (34). The increased fibrosis may also be an adaptive
response to reduced elasticity of the arterial wall in the area of the lesion. An earlier in vivo
study showed that elasticity in the area of stenosis can exacerbate the pressure drop across
the lesion, thereby reducing flow (35).

(A) There was no correlation between angiographic stenosis and tissue characterization (B). There was a
significant positive correlation between FFRmyo and fibrosis, and a significant negative correlation
between FFRmyo and the lipid pool: ▲, %CA = percent calcification area; □, %F = percent fibrotic area;
●, %LP = percent lipid pool area. (Source: J. Cardiol; 2010 ;55,296—302)
Fig. 10. Relationship between stenosis and tissue characterization
Our studies showed that angina patients with reduced FFRmyo are likely to have reduced
functional capacity, and that the tissue characteristics of coronary plaques in intermediate
lesions affect functional stenosis. According to these studies, high exercise capacity in
patients with stable angiographic stenosis is linked to the stability of coronary plaques.
Another report on coronary computed tomographic angiography indicated that coronary
342 Advances in the Diagnosis of Coronary Atherosclerosis

plaque characteristics were identified in patients with progressively reduced exercise times,
and that functional ischemia was more severe in those with mixed plaque. (36)
Thus, though coronary angiography is one of the most accurate methods for diagnosing
coronary artery disease, comprehensive assessment of health status is necessary for stable
angina patients with angiographic stenosis.

6. References
[1] Zir LM, Miller SW, Dinsmore RE, Gilbert JP, Harthorne JW. Interobserver variability in
coronary angiography. Circulation. 1976;53:627-32.
[2] DeRouen TA, Murray JA, Owen W. Variability in the analysis of coronary arteriograms.
Circulation. 1977;55:324-8.
[3] Kern MJ, de Bruyne B, Pijls NH. From research to clinical practice: current role of
intracoronary physiologically based decision making in the cardiac catheterization
laboratory. J Am Coll Cardiol. 1997;30:613-20.
[4] Tanaka S, Noda T, Segawa T, Minagawa T, Watanabe S, Minatoguchi S. Relationship
between functional exercise capacity and functional stenosis in patients with stable
angina and intermediate coronary stenosis. Circ J. 2009 ;73:2308-14.
[5] Pijls NH, De Bruyne B, Peels K, Van Der Voort PH, Bonnier HJ, Bartunek J Koolen JJ,
Koolen JJ. Measurement of fractional flow reserve to assess the functional severity
of coronary-artery stenoses. N Engl J Med. 1996;334:1703-8.
[6] Pijls NH, Van Gelder B, Van der Voort P, Peels K, Bracke FA, Bonnier HJ, el Gamal MI.
Fractional flow reserve. A useful index to evaluate the influence of an epicardial
coronary stenosis on myocardial blood flow; Circulation. 1995;92:3183-93.
[7] Tanaka S, Yokoyama C, Kawamura I, Takasugi N, Kubota T, Ushikoshi H, Aoyama T,
Kawasaki M, Nishigaki K, Takemura G, Minatoguchi S. Conservative medication
follow-up for over 20 years of a patient with ischemic heart disease after diagnosis
of chronic total occlusion of the 3 main coronary arteries. Circ J. 2008;72:1205-9.
[8] Albertal M, Regar E, Van Langenhove G, Carlier SG, Serrano P, Boersma E, Bruyne B, Di
Mario C, Piek J, Serruys PW; DEBATE Investigators. Flow velocity and predictors
of a suboptimal coronary flow velocity reserve after coronary balloon angioplasty.
Eur Heart J. 2002;23:133-8.
[9] Albertal M, Voskuil M, Piek JJ, de Bruyne B, Van Langenhove G, Kay PI, Costa MA,
Boersma E, Beijsterveldt T, Sousa JE, Belardi JA, Serruys PW; Doppler Endpoints
Balloon Angioplasty Trial Europe (DEBATE) II Study Group. Coronary flow
velocity reserve after percutaneous interventions is predictive of periprocedural
outcome. Circulation. 2002;105:1573-8.
[10] Handberg E, Johnson BD, Arant CB, Wessel TR, Kerensky RA, von Mering G, Olson
MB, Reis SE, Shaw L, Bairey Merz CN, Sharaf BL, Sopko G, Pepine CJ. Impaired
coronary vascular reactivity and functional capacity in women: results from the
NHLBI Women's Ischemia Syndrome Evaluation (WISE) Study;J Am Coll Cardiol.
2006 7;47: S44-9.
[11] Meuwissen M, Chamuleau SA, Siebes M, Schotborgh CE, Koch KT, de Winter RJ, Bax
M, de Jong A, Spaan JA, Piek JJ. Role of variability in microvascular resistance on
fractional flow reserve and coronary blood flow velocity reserve in intermediate
coronary lesions. Circulation. 2001;103:184-7.
[12] Matsuo H, Watanabe S, Yasuda S, Hirose T, Iwama M, Tanaka S, Yamaki T, Ono K,
Takahashi H, Segawa T, Matsuno Y, Minatoguchi S, Fujiwara H. Myocardial perfusion
during transient slow-flow in the patient with old vein graft intervention: assessment by
Novel Insights Into Stenosis on Coronary Angiography–Outline
of Functional Assessment of Stable Angina Patients with Angiographic Stenosis 343

serial measurement of pressure-derived fractional flow reserve and thermodilution-


derived coronary flow reserve. Catheter Cardiovasc Interv. 2003;60:392-8.
[13] De Bruyne B, Pijls NH, Smith L, Wievegg M, Heyndrickx GR. Coronary thermodilution
to assess flow reserve: experimental validation. Circulation. 2001;104:2003-6.
[14] Pijls NH, De Bruyne B, Smith L, Aarnoudse W, Barbato E, Bartunek J, Bech GJ, Van De
Vosse F. Coronary thermodilution to assess flow reserve: validation in humans.
Circulation. 2002;105:2482-6.
[15] Miller DD, Donohue TJ, Younis LT, Bach RG, Aguirre FV, Wittry MD, Goodgold HM,
Chaitman BR, Kern MJ. Correlation of pharmacological 99mTc-sestamibi myocardial
perfusion imaging with poststenotic coronary flow reserve in patients with
angiographically intermediate coronary artery stenoses. Circulation. 1994;89:2150-60.
[16] Bech GJ, De Bruyne B, Pijls NH, de Muinck ED, Hoorntje JC, Escaned J, Stella PR,
Boersma E, Bartunek J, Koolen JJ, Wijns W. Fractional flow reserve to determine the
appropriateness of angioplasty in moderate coronary stenosis: a randomized
trial;Circulation. 2001;103:2928-34.
[17] Pijls NH, van Schaardenburgh P, Manoharan G, Boersma E, Bech JW, van't Veer M, Bär
F, Hoorntje J, Koolen J, Wijns W, de Bruyne B. Percutaneous coronary intervention
of functionally nonsignificant stenosis: 5-year follow-up of the DEFER Study;J Am
Coll Cardiol. 2007;49:2105-11.
[18] Tanaka S, Noda T, Segawa T, Iwama M, Minagawa T, Watanabe S, Minatoguchi S. Relation
between functional stenosis and tissue characterization of intermediate coronary
plaques in patients with stable coronary heart disease. J Cardiol. 2010;55:296-302.
[19] Tonino PA, De Bruyne B, Pijls NH, Siebert U, Ikeno F, van' t Veer M, Klauss V,
Manoharan G, Engstrøm T, Oldroyd KG, Ver Lee PN, MacCarthy PA, Fearon WF;
FAME Study Investigators. Fractional flow reserve versus angiography for guiding
percutaneous coronary intervention. N Engl J Med. 2009 15;360:213-24.
[20] Gulati M, Pandey DK, Arnsdorf MF, Lauderdale DS, Thisted RA, Wicklund RH, Al-
Hani AJ, Black HR. Exercise capacity and the risk of death in women: the St James
Women Take Heart Project. Circulation. 2003;108:1554-9.
[21] Mora S, Redberg RF, Cui Y, Whiteman MK, Flaws JA, Sharrett AR, Blumenthal RS.
Ability of exercise testing to predict cardiovascular and all-cause death in
asymptomatic women: a 20-year follow-up of the lipid research clinics prevalence
study. JAMA. 2003;290:1600-7.
[22] Hlatky MA, Boineau RE, Higginbotham MB, Lee KL, Mark DB, Califf RM, Cobb FR, Pryor
DB. A brief self-administered questionnaire to determine functional capacity (the Duke
Activity Status Index). A brief self-administered questionnaire to determine functional
capacity (the Duke Activity Status Index). Am J Cardiol. 1989;64:651-4.
[23] Rankin SL, Briffa TG, Morton AR, Hung J. A specific activity questionnaire to measure
the functional capacity of cardiac patients. Am J Cardiol. 1996;77:1220-3.
[24] Myers J, Bader D, Madhavan R, Froelicher V. Validation of a specific activity
questionnaire to estimate exercise tolerance in patients referred for exercise testing.
Am Heart J. 2001;142:1041-6.
[25] Boden WE, O'Rourke RA, Teo KK, Hartigan PM, Maron DJ, Kostuk WJ, Knudtson M,
Dada M, Casperson P, Harris CL, Chaitman BR, Shaw L, Gosselin G, Nawaz S, Title
LM, Gau G, Blaustein AS, Booth DC, Bates ER, Spertus JA, Berman DS, Mancini GB,
Weintraub WS; COURAGE Trial Research Group. Optimal medical therapy with or
without PCI for stable coronary disease. N Engl J Med. 2007;356:1503-16.
[26] Weintraub WS, Spertus JA, Kolm P, Maron DJ, Zhang Z, Jurkovitz C, Zhang W,
Hartigan PM, Lewis C, Veledar E, Bowen J, Dunbar SB, Deaton C, Kaufman S,
344 Advances in the Diagnosis of Coronary Atherosclerosis

O'Rourke RA, Goeree R, Barnett PG, Teo KK, Boden WE; COURAGE Trial Research
Group, Mancini GB. Effect of PCI on quality of life in patients with stable coronary
disease;N Engl J Med. 2008;359:677-87.
[27] Hambrecht R, Walther C, Möbius-Winkler S, Gielen S, Linke A, Conradi K, Erbs S,
Kluge R, Kendziorra K, Sabri O, Sick P, Schuler G. Percutaneous coronary
angioplasty compared with exercise training in patients with stable coronary artery
disease: a randomized trial.Circulation. 2004;109:1371-8.
[28] Barmeyer A, Meinertz T. Anaerobic threshold and maximal oxygen uptake in patients
with coronary artery disease and stable angina before and after percutaneous
transluminal coronary angioplasty;Cardiology. 2002;98: 127-31.
[29] Fraker TD Jr, Fihn SD; 2002 Chronic Stable Angina Writing Committee; American College
of Cardiology; American Heart Association, Gibbons RJ, Abrams J, Chatterjee K,
Daley J, Deedwania PC, Douglas JS, Ferguson TB Jr, Gardin JM, O'Rourke RA,
Williams SV, Smith SC Jr, Jacobs AK, Adams CD, Anderson JL, Buller CE, Creager
MA, Ettinger SM, Halperin JL, Hunt SA, Krumholz HM, Kushner FG, Lytle BW,
Nishimura R, Page RL, Riegel B, Tarkington LG, Yancy CW. 2007 chronic angina
focused update of the ACC/AHA 2002 guidelines for the management of patients
with chronic stable angina: a report of the American College of Cardiology/American
Heart Association Task Force on Practice Guidelines Writing Group to develop the
focused update of the 2002 guidelines for the management of patients with chronic
stable angina.J Am Coll Cardiol. 2007;50:2264-74.
[30] Sano K, Kawasaki M, Ishihara Y, Okubo M, Tsuchiya K, Nishigaki K, Zhou X,
Minatoguchi S, Fujita H, Fujiwara H. Assessment of vulnerable plaques causing
acute coronary syndrome using integrated backscatter intravascular ultrasound. J
Am Coll Cardiol. 2006;47:734-41.
[31] Nissen SE, Yock P. Intravascular ultrasound: novel pathophysiological insights and
current clinical applications. Circulation. 2001;103:604-16.
[32] Kawasaki M, Takatsu H, Noda T, Sano K, Ito Y, Hayakawa K, Tsuchiya K, Arai M,
Nishigaki K, Takemura G, Minatoguchi S, Fujiwara T, Fujiwara H. In vivo
quantitative tissue characterization of human coronary arterial plaques by use of
integrated backscatter intravascular ultrasound and comparison with angioscopic
findings. Circulation. 2002;105:2487-92
[33] Okubo M, Kawasaki M, Ishihara Y, Takeyama U, Kubota T, Yamaki T, Ojio S, Nishigaki
K, Takemura G, Saio M, Takami T, Minatoguchi S, Fujiwara H. Development of
integrated backscatter intravascular ultrasound for tissue characterization of
coronary plaques. Ultrasound Med Biol. 2008;34:655-63.
[34] Stary HC, Chandler AB, Dinsmore RE, Fuster V, Glagov S, Insull W Jr, Rosenfeld ME,
Schwartz CJ, Wagner WD, Wissler RW. A definition of advanced types of
atherosclerotic lesions and a histological classification of atherosclerosis. A report
from the Committee on Vascular Lesions of the Council on Arteriosclerosis,
American Heart Association. Circulation. 1995;92:1355–1374.
[35] Siebes M, Campbell CS, D'Argenio DZ. Fluid dynamics of a partially collapsible stenosis
in a flow model of the coronary circulation. J Biomech Eng. 1996 ;118:489-97
[36] Lin FY, Saba S, Weinsaft JW, Wong FJ, Szulc M, Kligfeld P, Okin PM, Berman DS, Shaw
LJ, Min JK. Relation of plaque characteristics defined by coronary computed
tomographic angiography to ST-segment depression and impaired functional
capacity during exercise treadmill testing in patients suspected of having coronary
heart disease.Am J Cardiol. 2009 ;103:50-8.
19

Optimization of Radiation Dose


and Image Quality in Cardiac
Catheterization Laboratories
Octavian Dragusin1, Christina Bokou1,
Daniel Wagner2 and Jean Beissel2
1Entente des Hôpitaux Luxembourgeois
2Institut National de Chirurgie Cardiaque
et de Cardiologie Interventionnelle
Luxembourg

1. Introduction
Radiation exposure is a concern for those who participate in the practice of interventional
cardiology. However, many cardiologists are unaware that they may be exposing patients
to relatively high levels of radiation during cardiac catheterization procedures.
Interventional cardiology techniques are highly appreciated for its less invasive character
than surgery. A wide range of interventions, from diagnosis to treatment of heart and
coronary specific disorders are performed in both adults and children. Interventional
cardiology procedures can involve significant radiation exposure. Therefore, it is essential to
reduce the radiation dose as far as possible, by keeping an adequate image quality for
diagnostic and therapeutic purposes. Over the last years, introduction of new technologies
like flat-panel detectors have revolutionized fluoroscopy imaging. Most of the digital
detectors can potentially be used at lower doses. In practice, technical optimization studies
are rarely reported (especially for cardiac catheterization laboratories), notwithstanding the
fact that optimal settings could be very different from the optimal settings with the
traditional image intensifier systems.
This chapter is structured in three parts. The first part focuses on background notions
concerning X-ray production, radiation quantities and units, personal health risks and
concepts of radiation protection. The second part reviews the radiation exposure in
cardiovascular practice. The last part presents optimization strategies of image quality and
radiation dose and recommendations for limiting radiation exposures.

2. Radiation dose related to interventional cardiology


X-rays are special types of electromagnetic radiation which can ionize matter. The
interaction of radiation with tissue is dependent on the spectral distribution of the radiation
as well as on the thickness, density, and atomic composition of the matter. Cardiac imaging
systems use two forms of dynamic X-ray imaging: fluoroscopy and cineangiography.
Fluoroscopy mode uses relatively low radiation dose levels and aids in the guidance and
346 Advances in the Diagnosis of Coronary Atherosclerosis

positioning of medical devices within the patient. Cineangiography mode (shortly cine
mode) is the acquisition of a series of high definition dynamic images. It is mainly used to
image vessels injected with contrast media. The necessity of obtaining high contrast and
sharp edge definition with low noise images requires higher radiation dose rates. The basic
components of a fluoroscopic imaging chain are shown in Figure 1. The chain is composed
by an X-ray tube and generator, which are capable to produce a stable X-ray output over
long exposure times, a detector mounted opposite the X-ray tube, capable of dynamic
imaging, digital image processing and storage facilities, and a display system capable for
viewing real-time or recorded digital image series.

Fig. 1. Example of a fluoroscopic imaging chain equipped with image intensifier


X-ray detector is a significant component of the overall imaging chain. Two generations of
detector technology are present in the catheterization laboratories: image intensifier (II)
connected to a charged coupled device camera and flat-panel detectors (FD), also known as
active-matrix flat-panel imagers. Briefly, for II-based fluoroscopy systems, X-ray photons
exiting the patient are incident on the II’s input surface. The photons strike the CsI input
phosphor, absorbed and produce a large number of light photons. Then electrons are
produced from visible light photons and accelerated until they hit the output screen. From
the collision with output screen results light photons that are recorded by a video or cine
camera. Charges coupled device camera is used for direct digitalization of the image. The
process is illustrated in Figure 2, left. The FD detectors are classified into indirect and direct
detectors. Indirect detectors use a phosphor (scintillator) material that absorbs X-rays and
produces a proportionate number of light photons that subsequently interact with a
photodiode electrode on the TFT array (thin film transistor). A typical indirect FD detector is
shown in Figure 2, right. Direct FDs use a semiconductor material sandwiched between two
electrodes to absorb and convert the X-ray energy directly into ion pairs. Currently,
amorphous selenium (a-Se) is the only clinical choice.
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 347

Fig. 2. X-ray detector technology. On the left, image intensifier (II) technology. On the right,
flat-panel indirect detector (FD) technology.
There is a common consensus that FDs provides advantages in terms of image quality and
dose efficiency comparing with II technology. Advantages include image uniformity, no
geometrical distortions, no veiling glare and vignetting, wider dynamic range and better
ergonomics for patient accessibility. On the other hand, for FDs, system noise remains a
limiting factor and performance is rather limited for low exposure levels. (Holmes et at.,
2004; Seibert, 2006; Davies et al., 2007).

2.1 Radiation quantities and units


Quantities describing patient dose from radiation exposures have been defined by the
International Commission on Radiation Protection (ICRP, Publication 60, 1990 and ICRP,
Publication 103, 2007) and by the International Commission on Radiation Units and
Measurements (ICRU, Report 74, 2005). To make it simple, we will focus only on dose
quantities and radiation units that are often used in interventional cardiology. We propose
to divide them in three categories: dose quantities outside the patient’s body, dose quantities
to estimate risks of skin injuries and effects that have threshold and dose quantities to
estimate stochastic risks.
Radiation quantities outside the patient’s body are used to describe the beam of X-rays.
These quantities can express total amount of radiation or can express radiation at a specific
point. The X-ray beam is emitted from a small source (point) from the tube and is constantly
spreading out as it moves away from the source. Absorbed dose, D, is the mean energy
imparted per unit mass by ionizing radiation. The SI unit of D is the Gray (Gy), where 1 Gy
= 1 J/Kg. The Kerma (Kinetic Energy Released in a MAterial) is the sum of the initial kinetic
energies of all charged ionizing particles liberated by uncharged particles in a material of a
specific mass. The SI unit is also Gray (Gy). To make it simply, in diagnostic radiology,
absorbed dose D and Kerma are equal. It is possible to calculate the absorbed dose in a
material if the exposure is known, by using a conversion coefficient depending on medium.
Absorbed dose or air kerma in X-ray field can be measured with dosimeters. A practical
quantity that gives an indication of the absorbed dose is dose-area product (DAP) or kerma-
area product (KAP). It is the product of dose to air (air kerma) and the area of the X-ray
beam, and it is expressed in units Gy*cm². The use of DAP-meter provides a complete
348 Advances in the Diagnosis of Coronary Atherosclerosis

measurement of the total exposure of the patient. The DAP-meter is a flat large-area
ionization chamber mounted at the exit window of the X-ray tube and intercepts the entire
useful beam. In interventional cardiology, we work also with other quantities that influence
radiation dose. One of them is fluoroscopy time, which can be adopted as starting unit in a
quality assurance program (for comparison between operators, centers, procedures, for the
evaluation of protocols, and for the evaluation of operator skills). Other quantity is the
number of acquired cineangiographic images or number of series.
Entrance skin dose (ESD) includes the scatter from the patient. ESD = D*BF, where
backscatter factor ranges from 1.2-1.4 as a function of field size. The SI unit for ESD is the
Gray (Gy). Values of absorbed dose to tissue, compared with to air, will vary by a few
percent depending on the exact composition of the medium that is taken to represent soft
tissue. To evaluate the dose inside the patient’s body, we use the mean absorbed dose in a
tissue or organ, DT, in fact the energy deposited in the organ divided by the mass of that
organ. Organ doses cannot be measured on real patients. They can be measured in
anthropomorphic phantoms simulating examinations or by dedicated software tools that
simulates the interaction of X-rays on mathematical or realistic phantoms.

Fig. 3. Dosimetry terminology used in interventional cardiology (Adapted from ICRP


Publication 85).
A second category of dose quantities are the ones related to estimate risks of skin injuries
and effects that have threshold. Approximate entrance dose thresholds have been
determined for various skin changes, ranging from early transient erythema at 2 Gy to
painful dermal necrosis of about 18 Gy. For interventional procedures, maximal skin dose
(MSD) can be assessed with different methodologies: (a) by direct calculation, with off or
on-line techniques, (b) by direct measurements on the patient with point detectors
(thermoluminescent detectors, TLDs, or other solid state detectors), (c) by direct
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 349

measurements on the patient with large area detectors (films and TLDs array), and (d) by
portal monitoring with area or point/area detectors. The new cardiac X-ray systems are
able to display on the monitors dosimetric parameters at the interventional reference
point (IRP). The IRP is the reference point intended to be representative of the position of
the patient’s skin at the entrance site of the X-ray beam during an interventional
procedure. For fluoroscopic systems with an isocentre, the IRP is located along the central
ray of the X-ray beam at a distance of 15 cm from the isocentre in the direction of the focal
spot (Figure 3).
Radiation exposure of the different organs and tissues in the body results in different
probabilities of harm and different severity. The combination of probability and severity of
harm is called detriment. For instance in young patients, organ doses may significantly
increase the risk of radiation-induced cancer in later life. The equivalent dose (H) is the
absorbed dose multiplied by a dimensionless radiation weighting factor, wR, which
expresses the biological effectiveness of a given type of radiation. H  D  wR . The SI unit of
H is the Sievert (Sv). For X-rays, the wR is 1 so H is equal to D. The mean equivalent dose in
a tissue or organ HT is the energy deposited in the organ divided by the mass of that organ.
To reflect the detriment from stochastic effects due to the equivalent doses in the different
organs and tissues of the body, the equivalent dose is multiplied by a tissue weighting
factor, wT. The equivalent doses to organs and tissues weighted by the relative wT are
summed over the whole body to give the effective dose, E. The SI unit of E is also the Sievert
(Sv). E   T wT  HT . Till 2007 the tissue weighting factors of ICRP Publication 60 were
used. A recent estimation of those factors was published in ICRP report 103. Main
differences between publications for tissue weighting factors, wT are presented in Table 1.
Stochastic risk is calculated multiplying effective dose by a risk factor specific for sex and
age at the exposure.
Figure 3 illustrates the dosimetry terminology used in interventional cardiology. To
summarize, the biological effects and risks from ionizing radiation are divided into
deterministic and stochastic effects. Deterministic effects have a dose threshold, and the
intensity of the effect increases with increasing dose. Stochastic effects include cancer and
genetic risk. Probability of stochastic risks increases with increasing dose, but the intensity
of the effect is not a function of the absorbed dose. For personnel working in cardiac
catetherization laboratories, the major worries about radiation exposure are the potential
risk of cancer, cataracts, and genetic birth defects of pregnant staff if the appropriate
protective measures are not used. For patients, deterministic effects can be produced by
high doses of fluoroscopic X-rays. In addition, stochastic effects can appear on patients
with repeated interventional cardiac procedures, especially if are performed in young
population.
Risks of exposure to ionizing radiation are contra balanced by the need for and potential
benefit for a patient to have a cardiac interventional procedure. In fact medical use of
radiation is a unique situation in which patients are intentionally irradiated. The risk should
be minimised by utilizing techniques and procedures that keep exposure to a level As Low
As Reasonably Achievable (ALARA principle). The principle of ALARA is the axiom for all
radiation workers. Its successful implementation in catheterization laboratory require
understanding of factors responsible for levels of radiation exposure and applying
optimized procedures to obtain the medical diagnostic or therapeutic goal with minimum
radiation risks.
350 Advances in the Diagnosis of Coronary Atherosclerosis

ICRP Report 60 (1990) ICRP Report 103 (2007)


Tissue Tissue weighting factor, wT Tissue Tissue weighting factor, wT
Lung 0.12 Lung 0.12
Stomach Stomach
Colon Colon
Bone Marrow Bone
Marrow
Breast
Remainder
Gonads 0.20 Gonads 0.08
Thyroid 0.05 Thyroid 0.04
Oesophagus Oesophagus
Bladder Bladder
Liver Liver
Breast
Remainder
Bone surface Bone
surface
Skin Skin
Brain
Salivary
glands
Table 1. Differences between wT tissue weighting factors in ICRP Publication 60 and
Publication 103.

3. Literature review of patient and staff doses in clinical practice


Interventional cardiology procedures are usually fluoroscopy guided diagnostic and
therapeutic interventions. Therapeutic interventions are often complex procedures and
sometimes procedures are repeated for the same patient. In these situations patient high
radiation dose levels occur. The specialized literature presents a series of case reports
describing deterministic effects with growing incidence and more concern than stochastic
long term risks. International Commission on Radiological Protection and World Health
Organisation has requested attention to the skin dose problems of complicated radiological
interventions. In this context, patient dosimetry approaches are an important issue. Many
published dose data are expressed in terms of not particularly well defined dose quantities
and without clear objective for the measurements performed. This section of the work aims
to report and compare dosimetry approaches and eventually propose a classification of the
different dosimetry objectives. Patient dosimetry methods currently used in interventional
cardiology may be divided in three categories according to their purposes: (1) dosimetry for
quality assurance; (2) dosimetry for stochastic risk evaluation, (3) dosimetry for
deterministic effects evaluation.
Dosimetry for quality assurance is used to compare performance of equipment, operator
skills or radiological practice among different teams or centers, to evaluate the
optimization process or to establish diagnostic reference levels (DRL) for routine
examinations. Useful dose quantity is the dose – area product, DAP. For cardiac
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 351

procedures, usually the total DAP for whole procedure is documented. Sometimes, the
DAP for the fluoroscopy part (DAPfluoro) and the DAP for the cineangiography part of a
procedure (DAPcine) are indicated. Some authors propose additional technical quantities
like the total fluoroscopy time of a procedure, the total number of acquired images, the
number of series of acquired images, the mean number of images per series in a
procedure. Dosimetry quantities for stochastic risk evaluation are organ equivalent
doses to organ/tissue and effective dose. These quantities are indicators of overall
exposure in the assessment of stochastic effects of radiation exposure for population. Use
of these quantities allows us to compare exposures from different types of procedure,
radiation type, and radiation quality, and irradiation geometry. From effective dose, using
correct risk coefficients (ICRP Report 103), we can estimate the detriment of individuals or
population from medical exposure. Dosimetry for deterministic effects focuses on skin
dose assessment. For complex interventional cardiology procedures, the knowledge of
irradiated skin area is important with respect to the potential for deterministic effects of
radiation exposure. Different methodologies are used to express the maximal skin dose
(MSD): off-line or on-line calculation, direct measurements on the patient with point
detectors (TLDs or solid state detectors), direct measurements on the patient with the area
detectors (TLD array and film) or by portal monitoring with point or area detectors.
Unfortunately not all methods are feasible for routine work, specially the on-line ones. An
alternative approach on two levels can be used. The first level able to prevent
deterministic effects is using a threshold. This can be fluoroscopy time or a measurable
quantity like DAP, able to alert the operator that a certain value of skin dose,
corresponding to a threshold for deterministic effects can be reached. The second level to
assess MSD requires specific methods. In case of electrophysiology procedures where
stationary fields are used, electronic point detectors or computational methods able to
assess skin dose distribution can be employed.
We will present a summary of published patient doses in interventional cardiology. Any
attempt to compare published studies must be performed with circumspection due to the lack
of standardization of data acquisition and the uncontrolled variables (equipment differences,
radiographic technique, complexity of procedure, patient characteristics (size, age, sex).

3.1 Patient radiation doses during diagnostic and interventional cardiac


catheterization
The most frequent procedures in adults are coronary angiography (CA) for diagnostic and
percutaneous coronary intervention (PCI) for therapeutic examinations. Consequently, there
are a large number of articles that review the patient radiation doses. For CA procedures,
some authors report data for subcategories of procedures like CA with left ventricular
angiography, or/and cardiac catheterization. PCIs are more complicated to classify because
of the large number of technical factors, like number of lesions treated, the wire technique,
simple, ostial or bifurcation stenting, flow and pressure wire or any other special devices.
The most common quantities used were: DAP, fluoroscopy time, cine frames, cine time
(dosimetry for quality assurance), effective dose (dosimetry for stochastic risks), and skin
dose (dosimetry for deterministic risks). Published results are presented in Table 2 for CA
and in Table 3 for PCI procedures. The studies presented are a little part of the literature
survey. We present only median values and, where available, the range. We looked for
studies from different time periods. It is evident from the tabulated data that the reported
352 Advances in the Diagnosis of Coronary Atherosclerosis

values for interventional cardiology procedures vary considerably. This can be attributed to
procedural complexity, examination technique, use of radiation-reducing techniques,
operator experience, workload, and catheterization laboratory examination. For example,
Broadhead et al, 1997 compared patient dosimetry between single intensifier system and
biplane image intensifier system, and found that the biplane system provides greater
imaging capability but with increased dose. DAP for CA is 47.7 vs. 23.4 Gycm² (biplane vs.
single system), and for PCI is 72.2 vs. 51.6 Gycm².

Sample Maximum Effective


Fluoroscopy Cine frames
Study (number DAP (Gycm²) skin dose dose
time (min) (images)
cases) (mGy) (mSv)
Median ( Range min - max)
Zorzetto, 1997 52.5 4.9 1350
Broadhead, 1997 2174 58.8 9.4
van de Putte,
56.8 ( max 144) 412 – 725
2000
Eftathopoulos,
20 29 5
2003
Kuon, 2003 509 23.6 3.4 339
767
Dragusin, 2005 78 25.6 (9 -115) 4 (1 – 25)
(318 – 1331)
Karambatsakiou,
20 (18 -107)
2005
Vijakalakshmi,
3752 19.1 4.8
2007
D’Helft, 2008 13.6 – 231 0.22 – 27.6
688
Tsapaki, 2008 549 31 ( 1 – 135) 4 (0.8-57)
(62 -2206)
Bogaert, 2008 200 7.3
Dragusin, 2010 122 11.6 3 506
Samara, 2010 55 ( max 235) 3.1 ( max 65)
Table 2. Short review of patient doses during coronary angiography (CA) examinations
Bernardi et al., 2000 and Padovani et al., 2001 investigated the effect of complexity of PCI
interventions, by separating the procedures in “simple”, “medium” and “complex”
procedure, based on a set of technical and clinical factors. (Median DAP values 66.7,96.4 and
132.7 Gycm²). Sandborg et al, 2003 compared the radial arterial approach to the femoral
approach in term of radiation dose. They found that radial approach yielded significant
higher doses (51 and 75 Gycm² for CA, respectively PCI), compared with femoral approach
(38 and 47 Gycm²). Tsapaki et al. 2004 and Trianni et al., 2005a compared the patient doses
of flat-panel systems (FD) to image intensifier (II) systems. Despite of the potential of FD to
produce images of higher quality with lower entrance detector dose rates, in clinical practice
and terms of patient dose could give opposite results. Trianni et al., 2005a reported 33.4
Gycm² (for FD) and 31,1 Gycm² (for II) during CA examinations and 66.9 Gycm² (for FD)
and 52 Gycm² (for II) during PCI examinations. Some publications presented patient
dosimetry in term of contribution of fluoroscopy and image acquisition to the total DAP
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 353

(Efstathopoulos et al., 2003, Kuon et al., 2004). Mean DAP for fluoroscopy and image
acquisition calculated from the values reported in these studies are 18% DAP fluoroscopy
and 81% DAP image acquisition for CA procedures. For PCIs, DAP is 41% from fluoroscopy
and 59% from image acquisition. The patient radiation doses vary widely among published
studies. These differences are related to patient, procedure, X-ray equipment and physician.

Sample Maximum Effective


DAP Fluoroscopy Cine frames
Study (number skin dose dose
(Gycm²) time (min) (images)
cases) (mGy) (mSv)
Median ( Range min - max)
Zorzetto, 1997 82.6 12.2 1500
Broadhead,
214 77.9 14.2
1997
van de Putte,
108 – 131
2000
Eftathopoulos,
20 75 14
2003
Kuon, 2003 233 22.2 9.7 208
Dragusin, 2005 23 9 – 148.4 1.4 -44.5 206 -1524
Karambatsakiou,
10 16 - 115
2005
Vijakalakshmi,
646 35 9.3
2007
D’Helft, 2008 47.5 - 413 2-98
1257 799
Tsapaki, 2008 549 62 10.4 (3.2-53)
(398-5940) (320-1660)
Bogaert, 2008 118 11.6
Dragusin, 2010 91 32.7 12.6 938
Samara, 2010 144 9.1 (max 52)
Table 3. Short review of patient doses during percutaneous coronary intervention (PCI)
examinations

3.2 Patient radiation doses radiation doses during electrophysiology studies and
pacemaker implantations
Cardiac electrophysiology is a subdiscipline of interventional cardiology that is focused on
elucidating, diagnosing, and treating the electrical problems of the heart. The procedures are
performed by invasive intracardiac catheter recordings of spontaneous activity as well as of
cardiac responses to programmed electrical stimulation. These studies are performed to
evaluate abnormal electrocardiograms and to assess complex arrhythmias. Typically
therapeutic procedures are radiofrequency catheter ablations (RFCA). Patient doses are also
influenced by the type of procedures. Diagnostic electrophysiological study (EPS) relies
mainly on fluoroscopy. For therapeutic procedures, we can find different categories: RFCA
without angiographic images using contrast medium, RFCA with angiographic images
using contrast medium and RFCA of atrial fibrillation (AF) (Dragusin et al., 2005). The
dosimetric data of this study is presented in Table 4. In the case of therapeutic examinations,
354 Advances in the Diagnosis of Coronary Atherosclerosis

the radiation doses varied according to the location of the tachycardia. Ablations in which
contrast medium is used to take images need more radiation exposure. RFCA of atrial
fibrillation had longer examination times and yielded the maximum radiation doses.

Type of procedure Third


Parameter Range Mean ± SD Median
(number cases) quartile
EFO (72) 0.5 – 77.5 13.8 ± 16 7.2 17.3
DAP RFCA without contrast (85) 1.5 – 93.2 21.9 ± 20.5 14.3 27.9
(Gycm²) RFCA with contrast (52) 1.5 – 102.1 34.3 ± 26 32.1 47.6
AF (36) 33.6 – 440.2 156.3 ± 99.4 129.6 183.6
EFO (72) 1.6 – 39 11.2±8.6 8 13.8
Fluoroscopy RFCA without contrast (85) 4.4 – 48.8 23.7±11.2 20.7 30.3
time (min) RFCA with contrast (52) 10 – 61.3 26.5±11.2 22.9 31.2
AF (36) 27.6 - 130 67.6±24.7 66.4 85.5
Table 4. Summary of patient radiation data for electrophysiological procedures (Dragusin et
al., 2005)
Trianni et al., 2005b collected patient skin doses from 90 electrophysiological procedures.
Mean maximum local skin doses were 0.03 Gy for pacemaker insertion, 0.17 Gy for RFCA
for nodal tachycardia and 0.22 Gy for atrial fibrillation. Ector et al., 2007 investigated the
effect of body mass index on radiation doses in patients undergoing pulmonary vein
isolation for atrial fibrillation. 85 patients undergoing AF ablation guided by biplane low-
frequency pulsed fluoroscopy (3 frames/s). Mean DAP values per hour of fluoroscopy were
58, 110 and 184 Gycm² in normal, overweight, and obese patients. The corresponding
effective dose for AF ablation procedures were 15.2, 26.7, and 39 mSv.

3.3 Patient radiation doses radiation doses during pediatric cardiovascular


procedures
According to the Euratom Directive 97/43, exposures of children and procedures involving
high doses to the patient, such as interventional radiology, should be given special attention.
Both criteria apply to the pediatric cardiac catheterization laboratories, so patient dose
evaluations should thus be established with priority. On pediatric patients both diagnostic
and therapeutic procedures are performed. Diagnostic examinations study complex
congenital heart diseases. Therapeutic procedures mainly involve dilatation of stenotic
vessels or valves and occlusion of abnormal communications. The main therapeutic
procedures are balloon dilatation of the pulmonary valve, balloon dilatation of peripheral
pulmonary stenosis, balloon dilatation for coarctation of the aorta, stent implantation,
occlusion of patent ductus anteriosus, closure of atrial septal defect and closure of
ventricular septal defect. We will discuss the results of a study regarding pediatric radiation
doses performed in a catheterization laboratory equipped with a biplane FD system
(Dragusin et al., 2008a). In this study, the population was divided in six age groups (0-30
days, 1-12 months, 1-3 years, 3-5 years, 5-10 years and 10-15 years). Data from 273 patients
are presented in terms of DAP, fluoroscopy time and number of image acquisition series.
For diagnostic procedures the median values per age group are 2.7, 2.5, 5.1, 5.8, 7.1, 9.9
Gycm², 11, 6, 10, 8, 8, 5 minutes, and 8, 6, 6, 10, 8, 6 acquisition series. For therapeutic
procedures the median values per age group are 4.8, 5.9, 7.5, 9.5, 17.1, 46.8 Gycm² for DAP,
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 355

16.5, 12, 16, 18, 21, 20 minutes for fluoroscopy time and 9, 14, 12, 18, 22, 19 acquisition series.
The effective doses for each patient were estimated using PCXMC software (STUK, Finland).
The 25th, 50th and 75th percentile per each age group for diagnostic procedure is presented in
Figure 4 and for therapeutic procedures is presented in Figure 5.

Fig. 4. Estimated effective doses for diagnostic procedures in pediatric interventional


cardiology (Dragusin et al., 2008a)

Fig. 5. Estimated effective doses for therapeutic procedures in pediatric interventional


cardiology (Dragusin et al., 2008a)
Other study reported DAP values as 75th percentile of 6.2, 6.1, 9, 10, 15, 20, 27 and 36 Gycm²
for 2114 patients divided in eight age groups from newborns to 21 y (Rassow et al.,2000).
This study included diagnostic, therapeutic and myocardial biopsy procedures. Bacher et
al., 2005 studied 60 patients of which 28 underwent diagnostic imaging. Patients age ranged
from new born to 10 y. The DAP range for diagnostic was 0.96–14.6 Gycm2 and for
therapeutic 0.4–20.4 Gycm2. Onnasch et al.,2007 expressed the results of dose data of a large
population as DAP/body weight (Gycm²/kg) rather than DAPs for age groups. Using the
90th percentiles, he suggested as diagnostic reference level values of 0.81 Gycm²/kg for
diagnostic procedures, and 1.16 Gycm²/kg for therapeutic interventions. Effective doses of
0.6 to 23.2 mSv for diagnostic procedures and from 1 to 37 mSv for therapeutic procedures
were estimated by Bacher et al., 2005.
356 Advances in the Diagnosis of Coronary Atherosclerosis

A number of specific conditions, such as higher heart rates, smaller cardiovascular


structures, smaller body size, and wider variety of unusual anatomic variants with the
potential need for relatively lengthy and complex studies, result in relatively high radiation
doses to the patient. The improved survival of patients with complex anatomy (e.g.,
palliated single ventricle anatomies) implies that many such children with chronic cardiac
disease require frequent catheterizations within the first few years of life. These factors,
coupled with the increased radiosensitivity of children and a longer lifespan ahead of them
in which to possibly develop radiation-related sequels, converge to create potentially
unpleasant consequences.

3.4 Staff dosimetry


Occupational doses in interventional procedures guided by fluoroscopy are the highest
doses registered among medical staff using X-rays. Interventional cardiologists experience
radiation exposure with the patient, as they are close to the radiological source.
Interventional cardiologists working in high-volume cardiac catheterization laboratories are
exposed to significant occupational radiation risks of developing certain diseases, including
hematopoietic cancers, thyroid diseases, skin diseases, cataracts, or upper respiratory
disease (Venneri et al., 2009). Consequently, monitoring personnel exposure is compulsory
and regulated for individual workers. Occupational doses limits apply, i.e a limit on
effective dose of 20 mSv/year, averaged over 5 years. The equivalent dose should not
exceed 150 mSv for lens of the eye, 500 mSv for the skin (average dose over 1 cm² of the
most highly irradiated area of the skin) and 500 mSv for the hands and feet. Wraparound
two-piece aprons, thyroid shields and eye protection are important personnel shields.
Personnel dosimetry monitors include those using X-ray films (film badges) or
thermoluminescent dosimeters (TLDs). Both detectors are placed in holders and the
monitors are typically worn for one month before being submitted for processing. The
laboratory readout the detectors and estimate the effective dose. For interventional
cardiologists, it is recommended to wear two badges. One is worn outside the apron at the
neck and one is worn under the apron at the waist. The second badge monitors the
effectiveness of the lead apron. For other staff in the catheterization laboratory, a single
badge is worn and it is usually placed outside the apron at collar level. The mean collar-
level exposure per case for cardiologist who performs CA and PCI has been reported to be
0.04 to 0.16 mSv (Hujkens & Hummel, 1995, Zorzetto et al., 1997). Certain publications focus
on the estimation of personnel doses for different anatomical locations by placing TLDs for
each procedure. For example, Efstathopoulos et al, 2003 measured doses with five TLDs
placed on the left branch of eyeglasses, the chest over and underneath the lead apron, the
left hand and knee. The results of dose/procedure for cardiologists were 6 µGy (eyes), 0 µGy
(chest under apron), 5.75 µGy (chest over apron), 22.5 µGy (left hand), 16.75 µGy (left knee).
The assisting operator received less dose/procedure: 5.5 µGy (eyes), 0 µGy (chest under
apron), 4 µGy (chest over apron), 17.5 µGy (left hand), 3.5 µGy (left knee). An interesting
survey was performed by Vano et al, 2006, that evaluated the occupational doses of
cardiologist for a period of 15 years (1989 to 2004). The mean values in mSv/year decreased
from 11.6 (1989-1992) to 1.6 (1993-1998) and to 1.2 (1999-2004). The success of reduction in the
effective dose by a factor of 10 is explained mainly by the training in radiation protection,
optimization of procedures and improved performance of X-ray systems.
In conclusion, accumulation of radiation doses to interventional cardiologists has the
potential of increasing risk of stochastic effects. To prevent radiation–associated diseases,
radiation exposure to cardiologists should be reduced by using new generation cardiac X-
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 357

ray systems, performing diagnostic and interventional procedures with shorter fluoroscopic
times, and using effective shielding to protect from radiation.

4. Optimization strategies of image quality and radiation dose


In cardiac catheterization laboratory, the goal of the ALARA (As Low As Reasonably
Achievable) principle is to provide maximal diagnostic and therapeutic outcome while
requiring the lowest possible radiation dose. The following discussion focuses on the
compromise between radiation dose and image quality. There are few directions to follow.
First element on the equation of the dose-image quality is the cardiac catheterization
laboratory equipment. Taking into account the advances in technology, today modern
cardiac catheterization systems are equipped with reliable X-ray generation tubes and flat
panel detectors. If most of the laboratories are equipped with mono plane systems, for
electrophysiology or pediatric applications biplane equipments are probably more
appropriate. This choice depends on the cardiology practice. The imaging chain has an
important feature, the Automatic Exposure Control (AEC) that ensures relatively constant
image brightness. The AEC works as a feedback mechanism from the digital video processor
to the X-ray generator. As mentioned before, the cardiac X-ray system is capable of different
imaging modalities. Fluoroscopy mode is used for live, real-time viewing and provides
sufficient image quality to visualize the catheters. For permanent storage and review,
images of higher image quality are acquired in cineangiography mode. Nowadays, all
systems are delivered with pulsed fluoroscopy, with a range of pulse rate from 30 to 3
pulses/second. Note that the lower the pulse frequency the less radiation dose, at the
expense of a jerkier motion. The systems have the capability to store the last fluoroscopic
images as film (option “fluoro store” or “hold last image”). Cineangiography mode rates
vary from 60 to 15 or 30 frames/second. Most of adult angiograms are performed at 15
frames/second, but faster frame rates are necessary for pediatric patients to view rapidly
moving structures throughout the cardiac cycle.
Optimization of new cardiac equipment with flat panel detector consists of finding the
adapted configuration that offers an acceptable image quality with relatively low dose. The
detector entrance dose is known to influence the image quality, at least in the clinically used
dose range. In practice, various settings of the system lead to different patient dose levels.
The spectrum of the beam can be influenced by the kV, added Cu filtration, thickness of the
patient and the presence or absence of the grid. Dragusin et al., 2008b investigated influence
on image quality of three factors: detector entrance dose, effect of antiscatter grid and
patient thickness using two contrast phantoms (Leeds TO10 and CDRAD). These phantoms
were imaged in fluoroscopy and cineangiography mode. For cineangiography mode six
levels of detector entrance dose were used: 100, 120, 140, 170, 200 and 240 nGy/frame for a
rate of 15 frames/second. For fluoroscopy mode, three levels: low (10 pulses/second,
detector entrance dose 32 nGy/pulse), medium (15 pulses/second, 45 nGy/pulse) and high
(15 pulses/second, 65 nGy/pulse). The average fitted contrast-detail curve for CDRAD
phantom for different entrance detector doses is presented in Figure 6. The variation of the
contrast-detail curve with patient thickness (9 cm, 13 cm, 17 cm and 21 cm equivalent
plexiglass material) is presented in Figure 7. The configuration of X-ray system corresponds
to the acquisition mode 15 frames/second and 170 nGy/frame detector entrance dose.
When looking at graphically representation of contrast-detail curves, it should be noted that
improved image quality makes the curve to shift to the lower part of the graph. In Figure 6,
358 Advances in the Diagnosis of Coronary Atherosclerosis

it is evident that the poorest image quality corresponds to the fluoroscopy mode and the
better image quality corresponds to acquisition mode with maximum detector entrance dose
(240 nGy/frame).

10 45 nGy Fluoro 15 pps


100 nGy Cine 15fps
140 nGy Cine 15fps
200 nGy Cine 15fps
240 nGy Cine 15fps

0,1
0,1 1,0 10,0

Fig. 6. Contrast-detail curves for CDRAD phantom for fluoroscopy mode and cine mode for
different detector entrance doses.

10
9 cm
13 cm
17 cm
21 cm

0,1
0,1 1 10

Fig. 7. Contrast-detail curves for CDRAD phantom for cine mode (15 frames/second; 170
nGy/frame) and different simulated patient thicknesses.
The improvement in image quality between settings is not very clearly visible because of the
small changes in detector dose levels. Differences between detector dose settings are not
significan (a factor of 2.4 between the minimum 100 nGy and maximum 240 nGy). This
implies a little change in threshold contrast. Despite of the limitation of this study, the
authors observed that just a simple change of the detector entrance dose from a superior to a
next inferior setting (i.e. from 170 nGy/frame to 140 nGy/frame) will not dramatically
change image quality. This simple action would potentially reduce the patient skin dose
between 14-19% depending of the size of the patient. The limitation of the use of contrast
detail phantoms consists of the difficulty to link with clinical image quality requirements. In
particular for interventional cardiology, visualization of moving structures, visualization
and grading of subtle lesions or anatomical structures during the passage of a contrast agent
and the tracing of small catheters on the moving heart, implies the use of test objects that
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 359

simulates this environment. To visualize moving structures in cardiovascular fluoroscopy


systems, some dynamic phantoms have been created. Guibelalde et al., 2001 have
constructed the Patient Movement Simulation Test Object (PAMOSITO), a 2D motor-
controlled test object holder to simulate clinical situations in which patient movement could
be a cause of image degradation. The Society for Cardiac Angiography and Interventions
(SCA&I) and the National Electrical Manufacturers Association (NEMA) developed a
phantom that allows visualizing moving structures. The device is a rotating spoke that
contains five steel wires of different diameters. Two lead dots are used to evaluate lag and
recursive filtering (Balter et al.,2001). Both dynamic phantoms have the disadvantage that
they do not incorporate details or features that are directly linked with critical issues in
clinical cardiac images (i.e., circulation of contrast agent, anatomical background, lesions or
pulsating arteries).
Dragusin et al., 2008c used a phantom that models the human anatomy from the knee to the
neck. The modular design of the phantom contains also the heart (dimensions 15 cm
(atria/apex) x 10 cm wide x 9 cm (anterior/posterior). The arterial tree model is fabricated in
transparent polyurethane. Calcifications can be inserted into main arteries. Medical devices
like catheters can be inserted into the model, positioned in a particular arterial branch and
visualized on X-ray images. A pump circulates flow through the vasculature of the phantom.
The contrast agent boluses are injected in the circuit via femoral catheters (Figure 8).

Fig. 8. (Left) Synthetic Arterial Model (SAM) integrated into phantom body. (Right) Close-
up view of the heart model with coronary grooves highlighted
In this experiment were studied the effect of four technical variable of the X-ray equipment
with an anticipated influence on image quality: tube voltage (kV), additional Copper
filtration (mm Cu), detector entrance dose and dynamic density optimization (DDO). The
last variable is particular to Siemens cardiac catheterization systems equipped with flat
panel detector and is a image quality (post processing) parameter that works in real-time by
harmonizing the distribution of gray steps in the image. The X-ray filming of the phantom
allows the cardiologist to track the contrast agent circulation, to identify coronary lesions
and evaluate their visibility in rapport with anatomical structures. In this study four
calcified structures in the coronary arteries were used to simulate stenotic lesions. The first
lesion was located on the proximal left anterior descending (LAD) segment, the second on
the mid LAD segment, the third on the proximal circumflex segment (Cx) and the fourth on
the distal Cx segment. Typical radiographic images, with indication of the lesions, are
presented in Figure 9.
360 Advances in the Diagnosis of Coronary Atherosclerosis

Fig. 9. X-ray images of the heart phantom under different experimental conditions. L1 to L4
indicate the location of simulated lesions.
The contrast media (Iodine) was injected in the phantom using the hand technique and a 6F
catheter. 15 to 20 ml of iodine was used at the same flow rates and the image sequence is
started a few seconds after the injection. For full characterization of the lesions, sets of two
coronary angiograms from two viewing angle were recorded: RAO 30° and LAO
40°/CAUD20° (where RAO the right anterior oblique view, LAO the left anterior oblique
direction and CAUD the caudal inclination). The duration of the angiograms was 5 seconds
and was acquired at a rate of 15 frames/second. The angiograms were presented in a
random order to eight interventional cardiologists (4 experienced interventional cardiologist
and 4 residents). The observers were asked to subjectively assess the visibility of each
simulated lesion and to give an overall score for each angiogram by using a five-point scale:
1 = not visible; 2 = poor visibility; 3 = acceptable visibility; 4 = good visibility and 5 = very
good visibility. The evaluation of angiograms is presented in terms of image quality (IQ)
scores. For every angiogram entrance skin dose was measured and an effective dose was
estimated. Mean IQ scores ranged from 1.68 to 4.88. The highest IQ scores were obtained for
the angiograms acquired with tube potential 80 kV, no added Cu filters, DDO 60%, RAO
and LAO views and the highest entrance detector dose that has been used in the present
study, namely 170 nGy/frame. Radiation doses (entrance skin dose approximately 40 mGy
and effective dose of 1 mSv) were estimated for angiograms acquired at 15 frames/second,
detector field-of-view 20 cm, and a length of 5 s. The following parameters improved the IQ
factor significantly: a change in tube potential from 96 to 80 kV, detector entrance dose from
100 nGy/frame to 170 nGy/frame, the absence of Copper filtration. DDO variable which is a
post-processing parameter should be carefully evaluated because it alters the quality of the
images independently of radiation exposure settings. The SAM anthropomorphic phantom
has the advantage of visualization of stenotic lesions during the injection of a contrast agent
and using an anatomical background. In the future, this phantom could potentially bridge
the gap between physics tests and the clinical reality in the catheterization laboratory.
Future generation of phantoms should simulate the beating heart.
Optimization of X-ray equipment settings through experimental measurements is necessary,
mainly when it involves new equipment or technology. The acceptance of optimized
settings that affects image quality and radiation dose has to be accompanied by the training
of the user. Authors of this work have experience in this area. In our center in Luxembourg
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 361

(Institut National de Chirurgie Cardiaque et de Cardiologie Interventionnelle), we perform


continuously an internal audit related to radiation protection in clinical practice. Some
results of our experience were already published (Bokou et al., 2008; Dragusin et al., 2010).
All interventional cardiologists have taken part of two days continuous education course in
radiation protection. The course was practically a discussion forum between cardiologists,
radiation protection advisors and application specialist of X-ray equipment manufacturer,
with theoretical and practical sessions demonstrating the different factors influencing patient
and staff radiation doses. Before and after the course, a patient doses survey was performed.
During that period the catheterization laboratories were equipped with image intensifiers.
Later, the X-ray systems were upgraded and image intensifiers were replaced with new flat-
panel detectors. The effects of education in radiation protection and new technology are visible
on patient radiation doses (Figure 10). A decrease of 22% and 23% is noted for CA and,
combined CA and PCI procedures. With FD system, the interventional procedures are
performed with 6 pulses/second for fluoroscopy, compared with 15 pulses/second used on II
system. For FD system, the entrance detector dose is 29 nGy/pulse for fluoroscopy, and 100
nGy/frame for cineangiography mode (lowest option available in our systems).

70,0 66,1

CA CA+PCI
60,0

50,9
50,0
43,0
DAP (Gycm²)

40,0

30,0
21,9
20,0 17,0
14,8

10,0

0,0
II before course II after course FD - new system

Fig. 10. Mean DAP patient values with II before and after training course and with FD for
CA and PCI examinations.
Based on the survey of patient doses, we were able to establish reference levels (RL) for
common interventional procedures. These indicators are DAP, fluoroscopy time and
number of images, and give to the operator somehow the guideline of good and normal
practice. Of course, higher doses might occur if the clinical status of the patient justifies the
corresponding exposures to ionizing radiations. The goal to use RL is to control the level of
optimization of the procedures. Once the dose indicators are collected, the RL are
established as third quartile (75% percentile) of the distribution of observed data. In our
center, INCCI Luxembourg, local RL values for CA procedures are: 23 Gycm² for DAP, 5
minutes for fluoroscopy time and 617 images. For PCI procedures, the RL values are 44
Gycm² for DAP, 15.5 minutes for fluoroscopy time and 1163 images. Our values were
compared with reference levels at European level proposed by the SENTINEL consortium
(Padovani et al., 2008). The data was collected from nine European centers, 672 CA and 662
PCI procedures. Comparison of our local reference values with the European values are
shown in Figure 11 for CA procedures and in Figure 12 for PCI procedures.
362 Advances in the Diagnosis of Coronary Atherosclerosis

3rd quartile Local RL European RL


60

45

30

15

0
KAP [Gycm²] CF (x100) FT [min]

Fig. 11. Comparison of diagnostic reference levels for CA procedures between INCCI (local
RL) and European study

3rd quartile Local RL European RL


90

75

60

45

30

15

0
KAP [Gycm²] CF (x100) FT [min]
Fig. 12. Comparison of diagnostic reference levels for PCI procedures between INCCI (local
RL) and European study
Strictly comparison between both studies is difficult to perform, because we do not have
complete information about how the X-ray cardiac systems in the European study were
configured. Also, we do not know if the cardiologist’s experience and training education in
radiation protection is uniform between centers.
Probably one of the factors that explain the difference is the configuration of the X-ray
equipment for fluoroscopy mode. In our center the pulse rate is 6 pulses/s (low dose mode),
where in European study, centers that submitted data work usually with 12.5 frames/s (for
II systems) and 15 pulses/s (for FD systems). Our clinical experience shows that there is
possible to optimize protocols when using new technology with little compromise on image
quality but with benefit in reducing radiation dose levels to the patient.

5. Conclusion
Optimization of radiation dose - image quality and implementation of ALARA principle in
catheterization laboratories are tasks that involve all actors: interventional cardiologists,
auxiliary staff, medical physicist, and applications specialists. However, in daily practice
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 363

some tactics for radiation dose reduction and image quality improvement should be known
by all interventional cardiologists. We create a list of little steps that have an impact in
radiation safety in daily practice.
Use the lowest acceptable clinical protocols during fluoroscopy and cineangiography. Pulsed
fluoroscopy and cineangiography at lowest radiation level should be used. If the equipment
allows, different protocols must be created and used in function on the type of structure that
is being imaged (venous vs. arterial, fast-moving vs. slow-moving).
Correct placement of the patient in the isocenter on the table. Having the patient correct
positioned in the isocenter facilitates keeping the heart at the center of the X-ray field. In this
way there is no need of prolonged fluoroscopy to adjust the patient’s position with each
change in angiographic projection.
Avoid the use of fluoroscopy to make changes to the patient position or collimators. Fluoroscopy
should be used very briefly to check the patient position. Movements to the correct position
should be avoided by using fluoroscopy constantly. Modern units have “virtual” markers
that enable the positioning of the collimators. The correct position of collimators should be
checked by brief fluoroscopy rather then constant visualization.
Remove unnecessary instruments and body parts from the X-ray field. Presence of the patient’s
arm, operator’s hands or any external instruments should never be visible on a cardiac
study. These structures or objects result in an overall increase in radiation dose to the patient
because of the demand of AEC system to compensate with increased radiation output.
Minimize the number of angiograms. Limit the number of projections to provide an overview
of the status of the coronary arterial tree and indentify the ideal projections to be used for
coronary angioplasty. Always performs test injection of a small amount of contrast material
using fluoroscopy prior to acquiring an angiogram. This approach prevents the wasted
angiogram that is taken with the catheter inadvertently wedged deeply in a vessel. Also
fluoroscopy of test injection can aid in determining the correct magnification mode. Keep in
mind that few seconds of fluoroscopy and little quantity of contrast material are less
irradiating the patient than a full wasted angiogram. During complicated interventions,
limit the use of magnification, because of the substantial increase in radiation dose.
Keep the detector (II or FD) as close to the patient as possible. The X-ray tube should be as far
away as possible. If the detector is far from patient, the input doses will be higher and the
scatter radiation increases.
Decrease beam on time. This is probably one of the most important rules. Fluoroscopy must
not be applied when discussing or doing other manoeuvre. If the eye is not on the screen,
the foot should not be on the fluoroscopic pedal. Use stored images rather than live images
for studying the case.
Use angiographic projections that reduce operator exposure whenever possible. For right oblique
projections, the X-ray tube moves away from the operator, while for left anterior oblique
projection moves it closer. Kuon et al., 2004 published an interesting paper focused on
identification of less-irradiation tube angulations in invasive cardiology. Di Mario & Sutaria,
2005 published a review of techniques to obtain optimal views of all segments of the
coronary arterial system.
Remove anti-scatter grid when imaging small children. New cardiac X-ray systems have
possibility to remove the grid. In pediatric patients a significant reduction in radiation dose
is possible without compromising image quality.
Know your own cardiac X-ray equipment and its features. Work with radiation physicist, the
manufacturer to regularly test and maintain the equipment in optimal working conditions.
364 Advances in the Diagnosis of Coronary Atherosclerosis

Ensure protection of laboratory staff. Before starting fluoroscopy, ensure that everyone in the
room use radiation protection shielding. Ask of laboratory staff to keep distance during cine
angiography. Remember the inverse square law: doubling the distance from a point source
reduces the radiation exposure to one-quarter. Keep the proper use and storage of lead
aprons. Aprons that are not proper storage might develop cracks, compromising their
effectiveness. Always use personal dosimeters.
Technology of X-ray cardiac system is continuously adapting and optimization of radiation
doses with less compromise on image quality is possible. On the other side, activity in
cardiac catheterization laboratories increases and more complex procedures are performed.
All the actors involved in catheterization laboratory should be trained and familiar with the
basic principles of radiation safety. Developing a radiation safety culture should be a
priority. Attention to the simple rules of radiation safety and the planning of an
interventional procedure should enable the interventional cardiologist to produce high
quality images at low radiation level to the patient.

6. References
Bacher ,K; Bogaert, E; Lapere, R; De Wolf, D, &Thierens, H. (2005). Patient-specific dose and
radiation risk estimation in pediatric cardiac catheterization. Circulation.Vol 111,
pp.83-9.
Bernardi, G; Padovani, R; Morocutti, G; Vaño, E; Malisan, M.R; Rinuncini, M; Spedicato,
L; &Fioretti, P.M. (2000). Clinical and technical determinants of the complexity of
percutaneous transluminal coronary angioplasty procedures: analysis in relation to
radiation exposure parameters.Catheter Cardiovascular Interventional. Vol 1, pp.121-9
Bogaert, E; Bacher, K & Thierens, H (2008) A large-scale multicentre study in Belgium of
dose area product values and effective doses interventional cardiology using
contemporary X-ray equipment. Radiation Protection Dosimetry. Vol.128, pp.312-323
Broadhead, DA; Chapple, CL; Faulkner, K; Davies, ML; &McCallum, H;(1997). The impact
of cardiology on the collective effective dose in the North of England. British Journal
of Radiology, Vol 70, pp.492-7
Davies, G.A.; Cowen, A.R.; Kengyelics, S.M.; Moore, J. & Sivananthan, M.U. (2007). Do flat
detector cardiac X-ray systems convey advantages over image-intensifier-based
systems? Study comparing X-ray dose and image quality. European Radiology,
Vol.17, pp.1787-1794
D’Helft, C; McGee, A; Rainford, L; McFadden, S; Winder, J; Hughes, C; & Brennan, PC.
(2008) Proposed preliminary diagnostic reference levels for three common
interventional cardiology procedures in Ireland. Radiation Protection Dosimetry.
Vol.129, pp.63-6
Di Mario, C & Sutaria, N. (2005). Coronary angiography in the angioplasty era: projections
with a meaning. Heart. Vol. 91, pp. 968-976
Dragusin, O; Desmet , W ; Heidbuchel, H; Padovani, R; & Bosmans, H. (2005). Radiation
dose levels during interventional cardiology procedures in a tertiary care hospital.
Radiation Protection Dosimetry. Vol.117, pp.231-5.
Dragusin, O; Gewillig, W; Desmet, W; Smans, K; Struelens, L & Bosmans, H (2008a).
Radiation dose survey in a paediatric cardiac catheterization laboratory equipped
with flat-panel detectors Radiation Protection Dosimetry, Vol.129, pp.91-95
Optimization of Radiation Dose and Image Quality in Cardiac Catheterization Laboratories 365

Dragusin, O; Smans, K; Jacobs, J; Inal, T & Bosmans, H (2008b). Evaluation of the contrast-
detail response of a cardiovascular angiography system and the influence of
equipment variables on image quality. Proceedings SPIE Medical Imaging, Vol.6913,
San Diego, USA, March 2008
Dragusin, O; Bosmans, H; Pappas, C & Desmet, W. (2008c). An investigation of flat panel
equipment variables on image quality with a dedicated cardiac phantom, Physics in
Medicine and Biology, Vol.53, 4927-4940
Dragusin, O; Breisch, R; Bokou, C; & Beissel, J (2010). Does a flat panel detector reduce the
patient radiation dose in interventional cardiology? Radiation Protection Dosimetry,
Vol.139, pp.266-270
Ector, J; Dragusin, O; Adriaenssens, B; Huybrechts, W; Willems, R; Ector, H; & Heidbuchel,
H. (2007). Obesity is a major determinant of radiation dose in patients undergoing
pulmonary vein isolation for atrial fibrillation. Journal of the American College of
Cardiology. Vol 50, pp.234-242.
Efstathopoulos, EP; Makrygiannis, SS; Kottou, S; Karvouni, E; Giazitzoglou, E; Korovesis,
S; Tzanalaridou, E; Raptou, P.D; &Katritsis, D.G.(2003). Medical personnel and
patient dosimetry during coronary angiography and intervention. Physics in
Medecine and Biology,Vol 18, pp.3059-68.
Holmes, D.R.Jr.; Laskey, W.K.; Wondrow, M.A. & Cusma, J.T. (2004). Flat-panel detectors in
cardiac catheterization laboratory: revolution or evolution – what are the issues?
Catheterization and Cardiovascular Interventions, Vol.63, pp.324-330
Huyskens, CJ & Hummel, WA (1995). Data analysis on patient exposures in cardiac
angiography. Radiation Protection Dosimetry. Vol 57, pp. 475-480.
ICRP, 1990 Recommendations of the International Commission on Radiological Protection.
Publication 60. Ann. ICRP, Oxford, UK: Pergamon Press, 1991, 21(1-3)
ICRP, Avoidance of Radiation Injuries from Medical Interventional Procedures. Publication 85.
Ann. ICRP, Oxford, UK: Pergamon Press, 2000, 30
ICRP,The 2007 Recommendations of the International Commission on Radiological Protection.
Publication 103. Ann. ICRP, Oxford, UK: Pergamon Press, 2007, 37(2-4)
ICRU, International Commission on Radiation Units and Measurements. Patient dosimetry for
X-rays used in medical imaging. ICRU Report 74, 2005, Journal of ICRU, Vol.5, No.2
Karambatsakidou, A; Tornvall, P; Saleh ,N; Chouliaras, T; Löfberg, PO;& Fransson A.(2005).
Skin dose alarm levels in cardiac angiography procedures: is a single DAP value
sufficient? British Journal of Radiology,Vol 78,pp.803-9.
Kuon, E ; Glaser, C ; & Dahm, JB.(2003). Effective techniques for reduction of radiation
dosage to patients undergoing invasive cardiac procedures.British Journal of
Radiology.Vol 76, pp.406-13.
Kuon, E; Empen, K; Rohde, D; & Dahm, JB.(2004a). Radiation exposure to patients
undergoing percutaneous coronary interventions: are current reference values too
high? Herz,Vol 2, pp.208-17.
Kuon, E; Dahm, J; Empen, K; Robinson, D; Reuter, G & Wucherer, M. (2004b). Identification
of less-irradiation tube angulations in invasive cardiology. Journal of American
College of Cardiology. ,Vol 44, pp.1420-1428.
Onnasch, D. G. W.; Schroder, F. K.; Fischer, G; & Kramer, H. M. (2007). Diagnostic reference
levels and effective dose in paediatric cardiac catheterization. British Journal of
Radiology. Vol80, pp.185-177.
366 Advances in the Diagnosis of Coronary Atherosclerosis

Padovani, R; Bernardi, G; Malisan, M.R; Vañó, E; Morocutti, G, &Fioretti, P.M.(2001). Patient


dose related to the complexity of interventional cardiology procedures.Radiation
Protection Radiology.Vol 94, pp.189-92
Padovani, R; Vano, E; Trianni, A; Bokou, C; Bosmans, H; Bor, D; Jankowski, J; Torbica, P;
Kepler, K; Dowling, A; Milu, C; Tsapaki, V; Salat, D; Vassileva, J & Faulkner, K.
(2008). Reference levels at European level for cardiac interventional procedures.
Radiation Protection Dosimetry. Vol 129,pp. 104-107
Rassow, J; Schmaltz, A. Hentrich, F; & Streffer, C. (2000). Effective dose to patients from
paediatric cardiac catheterization. British Journal of Radiology.Vol 73,pp. 183-172.
Samara, TE; Aroua, A; Stauffer, JC; Bochud, F & Verdun, FR. (2010) Fluoroscopy-guided
procedures in cardiology:is patient exposure being reduced over time? Radiation
Protection Dosimetry, Vol.139, pp.271-274
Sandborg , M; Fransson, SG; & Pettersson, H. (2003). Evaluation of patient-absorbed doses
during coronary angiography and intervention by femoral and radial artery access.
European Journal of Radiology.Vol 4, pp.653-8.
Seibert, J.A. (2006). Flat-panel detectors: how much better are they? Pediatric Radiology,
Vol.36, pp.173-181
Trianni, A; Bernardi, G; & Padovani, R.(2005a). Are new technologies always reducing patient
doses in cardiac procedures? Radiation Protection Radiology.Vol 117, pp.101-97.
Trianni, A; Chizzola, G; Toh, H; Quai, E; Cragnolini, E; Bernardi, G; Proclemer, A &
Padovani, R.(2005b). Patient skin dosimetry in haemodynamic and
electrophysiology interventional cardiology. Radiation Protection Radiology.Vol 117,
pp.241-6.
Tsapaki, V; Kottou, S; Kollaros, N; Dafnomili, P; Kyriakidis, Z; &Neofotistou ,V.(2004). Dose
performance evaluation of a charge coupled device and a flat-panel digital
fluoroscopy system recently installed in an interventional cardiology laboratory.
Radiation Protection Radiology,Vol 3,pp.297-304.
Tsapaki, V; Patsilinakos, S; Voudris, V; Magginas,A; Pavlidis, S; Maounis, T; Theodorakis, G;
Koutelou, M; Vrantza, T; Nearchou, M; Nikolaki, N; Kollaros, N; Kyrozi, E; Kottou, S;
Karaiskos, P; Neofotistou, E; & Cokkinos, D. (2008) Level of patient and operator dose
in the largest cardiac centre in Greece. Radiation Protection Dosimetry. Vol.129, pp.71-3
Van de Putte, S; Verhaegen, F; Taeymans ,Y; &Thierens, H.(2000). Correlation of patient skin
doses in cardiac interventional radiology with dose-area product. British Journal of
Radiology, Vol 73, pp. 504-13.
Vano, E; Gonzalez, L; Fernandez, JM; Alfonso, F & Macaya, C. (2006). Occupational radiation
doses in interventional cardiology. British Journal of Radiology. Vol 79,pp. 383-8.
Venneri, L; Rossi, F; Botto, N; Grazia, M ; Salcone, N ; Emad, A ; Lazzeri, M ; Gori, C ; Vano,
E & Picano, E (2009). Cancer risk from professional exposure in staff working in
cardiac catheterization laboratory: Insights from the National Research Council’s
Biological Effects of Ionizing Radiation VII Report. The American Heart Journal. Vol
157, pp. 118-127.
Vijayalakshmi, K; Kelly, D ; Chapple, C.L; Williams, D ,Wright, R; Stewart, M.J; Hall, J.A;
Sutton, A;Davis, A; Haywood, J; & Belder, M.A.(2007).Cardiac catheterization:
radiation doses and lifetime risk of malignancy. Heart. Vol371, pp.370-371.
Zorzetto, M; Bernardi, G; Morocutti, G & Fontanelli, A. (1997) Radiation exposure to
patients and operators during diagnostic catheterization and coronary angioplasty,
Catheterization and cardiovascular diagnosis. Vol 40, pp. 348-51
20

Protection of the Patient and the Staff from


Radiation Exposure During Fluoroscopy-
Guided Procedures in Cardiology
Verdun Francis R., Aroua Abbas, Samara Eleni,
Bochud François and Stauffer Jean-François
Lausanne University Hospital
Switzerland

1. Introduction
The volume of diagnostic or therapeutic procedures in cardiology that require the use of
ionizing radiation is increasing constantly. Currently, technological developments offer the
possibility of exploring not only the cardiac function (measurement of ejection fraction, for
example) but also the state of coronary and great vessels. In fact, the management of
patients with heart disease often requires the use of investigative techniques using X-rays.
For example 3.85 million cardiac catheterizations were performed in the United States in
2002 (Einstein et al., 2007). In Switzerland the last national survey on the exposure of the
population by medical radiology revealed that nearly 34 000 coronary angiographies and
more than 18 000 coronary dilatations were performed in 2008. They are associated to 65%
of the collective dose related to interventional radiology and 8% to that related to all medical
X-rays (Samara et al. 2011).
The increase in radiological examinations using ionizing radiation has been mentioned for
several years not only in medical journals for professionals but also in the press addressing
the general public. For example, in its edition of 17 June, 2007, the New York Times
questioned the public opinion about the justification of the increasing number of CT
examinations.
While most examinations deliver relatively low doses and thus add only a low risk to the
procedure itself, there are situations where doses exceed the dose level where an excess risk
of death from cancer has been demonstrated. In addition, some complex procedures may
result in the occurrence of deterministic effects such as burns to the skin.
The substantial increase of fluoroscopy-guided procedures in cardiology over the past few
years has been accompanied by a parallel growth in concern for patient radiation safety and
for the safety of the operators who perform these procedures. Thus, radiation safety has
become a major issue in radiology departments.
The aim of this chapter is:
- to recall the effects of ionizing radiation on the human body and the radiological risks;
- to introduce the dosimetric quantities (basic and operational) commonly used to
quantify those risks;
- to briefly present the principles of radiation protection;
- to provide the tools (actions and means) necessary for operational radiation protection.
368 Advances in the Diagnosis of Coronary Atherosclerosis

2. Radiation effects and dose


The aim of this section is to recall the effects of ionizing radiation on the human body and
the radiological risks

2.1 Radiation effects


Effects associated with exposure to radiation are divided into two categories: stochastic and
deterministic.

2.1.1 Stochastic effects of radiations


The major risks associated with a radiological procedure using ionizing radiation are due to
stochastic effects. They induce a minimal genetic risk on offspring and especially add to the
natural risk of developing cancer an additional risk. They strongly depend on age and are
three to four times higher in children than in adults. One of the problems related to
stochastic effects is that they are considered to be without threshold. It is therefore necessary
to justify an examination using ionizing radiation and to be sure that the patient will benefit
from the radiological procedure exceeds the associated risk. In addition, when the
procedure is justified, the management of the patient dose must be optimized.

2.1.2 Deterministic effects of radiations


Deterministic effects include cataract, skin erythema, epilation, and skin burns at different
stages depending on the degree of irradiation of the skin. Unlike the stochastic effects,
deterministic effects present a threshold and appear only beyond a certain level of exposure
of the skin to radiation. They occur only a few days or months after irradiation and the
higher the dose to the skin is, the higher deterministic effects are severe as shown in table 1.
Deterministic effects are generally not expected to result from purely diagnostic cardiac
investigations, but are sometimes reported with the management of critical cases in
particular after some complex interventional procedures such as the dilatation of the
coronary arteries (PTCA), recanalizations or thermal ablations. They are certainly rare but
they do exist (Suzuki et al., 2008).

Effect Threshold dose (Gy) Onset


Early transient erythema 2 Few hours
Temporary epilation 3 3 weeks
Main erythema 6 10 days
Permanent epilation 7 3 weeks
Dry desquamation 10 4 weeks
Dermal atrophy 11 > 14 weeks
Telangiectasia 12 > 52 weeks
Moist desquamation 15 4 weeks
Late erythema 15 6 to 10 weeks
Necrosis 18 > 10 weeks
Table 1. Deterministic effects to the skin
Protection of the Patient and the Staff from Radiation
Exposure During Fluoroscopy-Guided Procedures in Cardiology 369

2.2 Basic dosimetric quantities


There are several dosimetric quantities that have been introduced to assess risks and control
the exposure of the patient and the staff. This section will briefly describe the fundamental
quantities.

2.2.1 Absorbed dose, D


X-rays or gamma rays are indirectly ionizing radiation because energy is released into the
tissue through the electrons set in motion by the X-rays or gamma rays, which in turn will
make a very large number of ionizations. The energy that these electrons deposit per unit
mass of tissue, T, or organ is called the absorbed dose and is denoted DT. This is the basic
physical quantity used to measure the biological effects expected. It has the dimension of
one joule per kilogram (J kg-1) and is expressed in gray (Gy). This quantity is used to control
the deterministic effects with a threshold of 0.5 Gy.

2.2.2 Equivalent dose, H


To reflect the fact that all types of radiation for a given absorbed dose, do not produce the
same effect in humans the concept of dose equivalent in a tissue, T, or organ, denoted HT
was introduced. It is the product of DT and a weighting factor, wR, which depends on the
type of radiation and expresses its effectiveness.

HT = Σ R wR . DT,R
HT has the same dimension as DT (J kg-1), but is expressed in sievert (Sv).
As shown in table 2, X-rays or gamma rays are taken as reference radiation and wR is
therefore equal to unity. Thus, an adsorbed dose of 1 mGy is equivalent to a dose equivalent
of 1 mSv

 Radiation type wR (ICRP60) wR (ICRP103)


 Photons, electrons, muons 1 1
 Protons, charged particles 5 2
 Alpha particles, heavy 20 20
nuclei, fission fragments
 Neutrons 5/10/20, depending on A continuous function of
energy neutron energy
Table 2. Radiation weighting factors according to ICRP60 and ICRP103

2.2.3 Effective dose, E


The effective dose (E) was firstly proposed by Jacobi in C). The aim of the E was to define a
quantity that could be directly related to the probability of a detriment from low-dose
exposure to ionizing radiation where only stochastic effects occur. The E concept was
adopted by ICRP in 1977 (ICRP26, 1977) and further developed in its Recommendations
ICRP60 (ICRP90, 1990) and ICRP103 (ICRP, 2007).
E is defined by the weighed sum of mean tissue and organ doses with radiation weighting
factors taking into account a) the different radio-biological effectiveness of various
radiations and b) the different sensitivity of tissue and organs with respect to stochastic
effects. E is defined as:
370 Advances in the Diagnosis of Coronary Atherosclerosis

E = Σ T wT . HT = Σ T,R wT . wR . DT,R
where wT is the tissue weighting factor (see table 3).

 Organ/Tissue ICRP60 ICRP103 ICRP103/ICRP60


 Gonads 0.20 0.08 0.4
 Colon 0.12 0.12 1.0
 Lungs 0.12 0.12 1.0
 Red bone marrow 0.12 0.12 1.0
 Stomach 0.12 0.12 1.0
 Bladder 0.05 0.04 0.8
 Breast 0.05 0.12 2.4
 Liver 0.05 0.04 0.8
 Oesophagus 0.05 0.04 0.8
 Thyroid 0.05 0.04 0.8
 Bone surface 0.01 0.01 1.0
 Skin 0.01 0.01 1.0
 Brain 0.01
 Salivary glands 0.01
 Remainder 0.05* 0.01+
*ICRP60 remainder tissues/organs: adrenals, brain, kidneys, muscle, pancreas, small intestine, large
intestine, spleen, thymus, uterus.
+ ICRP103 remainder tissues/organs: adrenals, extrathoracic tissue, gall bladder, heart, kidneys,

lymphatic nodes, muscle, oral mucosa, pancreas, prostate, small intestine, spleen, thymus,
uterus/cervix.
Table 3. Tissue weighting factors

2.3 Operational dosimetric quantities


As above indicated to account for stochastic effects the effective dose (E) has to be
determined. To estimate E associated with a radiological examination various operational
quantities, easy to measure, are used. They are also listed at the end of the examination on
the console of the X-ray unit

2.3.1 Entrance dose and dose area product


For radiographic examinations, the operational quantity used is called absorbed dose at the
surface at the entrance of the beam in the patient and abbreviated as ESD. The ESD is
expressed in mGy and is converted into effective dose, in mSv, by multiplying it by a factor
which is for example 0.2 for a chest radiograph.
For fluoroscopy procedures the operational quantity used is called dose-area product and
abbreviated as DAP. The unit commonly used is Gy.cm2. Some manufacturers also express
the DAP in cGy.cm2 or μGy.m2. To convert the DAP in Gy.cm2 into E in mSv, it has to be
multiplied by a converting factor equal to 0.2 in the case of the exposure of the chest region
(when Gy.cm2 is used).
Table 4 shows the operational quantity and the effective dose associated with a series of
routine examinations in diagnostic radiology.
Protection of the Patient and the Staff from Radiation
Exposure During Fluoroscopy-Guided Procedures in Cardiology 371

 Examination Operational quantity Typical values Effective dose (mSv)


 Bite-Wing (dental) ESD (mGy) 2 0.01
 OPG (dental) ESD (mGy) 0.7 0.06
 Chest PA ESD (mGy) 0.1 0.02
 Hip AP ESD (mGy) 6 0.7
 Abdomen PA ESD (mGy) 3 1.2
 Coronarography DAP (Gy.cm2) 60 12
 PCI DAP (Gy.cm2) 80 16
 RFCA DAP (Gy.cm2) 130 26
 CTCA DLP (Gy.cm) 1500 25
Table 4. Operational dose quantity and effective dose for a number of X-ray examinations

2.3.2 Skin accumulated dose


The estimation of absorbed dose at the surface of the skin by fluoroscopy, to account for
deterministic effects, is a difficult task since the examination is conducted with changing
regularly the incidence X-ray beam. However, it is possible to have an idea of the dose to the
skin using a particular operational quantity, called the cumulative dose. This quantity
estimates the dose that would have received the skin if the geometry was kept unchanged
throughout the procedure. It may be considered that the cumulative dose indicated by the
facility in cardiology overestimates the dose to the skin by a factor of 2-3 since several tube-
detector incidences are used that distributes the exposure of the skin. If this operational
quantity is not available, the skin dose could be estimated from the DAP knowing that we
will be much less accurate. To estimate the skin dose in Gy, in a situation where the tube
does not rotated around the patient during the procedure one can divide the DAP (Gy.cm2)
by 100 for a well collimated X-ray beam (100 cm2 : area of the X-ray beam at the entrance of
the patient).

2.3.3 Ambient dose equivalent


Another operational quantity used to assess the effective dose delivered to the operator is
the ambient dose equivalent, H*(10), at the point of interest in the actual radiation field. It is
defined (ICRU, 1992) as the dose equivalent which would be generated in the associated
oriented and expanded radiation field at a depth of 10 mm on the radius of the ICRU sphere
which is oriented opposite to the direction of incident radiation. This quantity is normalized
traceable. It is representative of the effective dose the staff receives.

3. Principle of radiation protection


Radiation protection is assured by respecting three general principles which are: the
justification of the practice, the optimisation of the protection and the individual dose
limits. The justification of the practice is due to the fact that exposure to ionising radiation
has deleterious effects on health. The principle of optimisation is introduced since some of
these effects are considered as non-threshold’s ones, and thus one has to reduce exposure
to levels. The principle of individual limits is introduced as a safeguard to prevent
situations where the respect of the two first principles would not be sufficient to protect
individuals.
372 Advances in the Diagnosis of Coronary Atherosclerosis

3.1 Justification
The first principle of radiation protection is justification. No exposure is acceptable unless its
usefulness is demonstrated. In the medical practice any radiological modality or procedure
has to be justified. It should be properly indicated to the diagnostic or therapeutic case. In
other words it should be demonstrated that no other non-irradiating non invasive modality
can give the same diagnostic or therapeutic results. In order to implement the justification
principle, learned society in several countries (USA, UK, France, Switzerland and others)
worked out referral guides in order to indicate which radiological modality/examination is
suited to which diagnostic or therapeutic case.

3.2 Optimization
The second principle of radiation protection is optimization. When the exposure to radiation
is justified, an effort must be engaged in order to keep the radiation dose delivered as low as
reasonably achievable (ALARA principle). If justification reflects the will “to do the right
thing”, optimisation reflects the will “to do it right”. In the medical practice the principle of
optimization is implemented in different ways contributing together to lower the radiation
doses to the patients and to the staff. The quality control of the X-ray unit is important to
make sure the optimal settings are used. The auditing of the protocols used for each
radiological procedure allows producing the quantity and quality of images sufficient to the
diagnostic or therapeutic goal, with an optimum trade-off between image quality and
radiation dose, and no unnecessary irradiation of the patient and staff. The use of protecting
means contribute to cut down the radiation (direct or scattered) that is not useful in the
imaging process. Training, informing, using diagnostic reference levels, all this helps spread
a culture of optimization.

3.3 Limitation
The third principle of radiation protection is dose limitation in order to avoid excessive
exposure of an individual. The ICRP recommends a set of dose limits for the general public
and for people exposed in the course of their occupation (ICRP 103, 2007). The principle of
dose limitation applies to health professionals (physicians, radiographers, medical
physicists, etc.) but it does not apply to the patient. The sound benefit (diagnostic or
therapeutic) from X-rays use implies that higher doses of radiation are tolerated as long as
the radiological procedure is justified and optimized.

4. Operational radiation protection


Near a radiological room radiation protection of workers and of public is ensured by the
limitation of the weekly ambient equivalent dose through the shielding of the rooms.
Surveys have shown that doses to public and workers in such an environment are very low.
However there is not ambient equivalent dose limitation near the X-ray imaging unit. Thus,
it is mandatory to wear a lead apron when being near a running X-ray imaging device.
Many professionals that use fluoroscopy units receive low dose of radiation since they only
use these unit a few minutes (or less) per patient. Unfortunately, this is often not the case for
cardiologists who can handle very complex procedures that not only expose the patient but
also expose the staff present in the suite. Thus an effort to improve the radiation protection
should get a high priority.
Protection of the Patient and the Staff from Radiation
Exposure During Fluoroscopy-Guided Procedures in Cardiology 373

It is worth mentioning that the exposure of staff is not due to X-ray leakage from the X-ray
tube of the fluoroscopy unit. Exposure to the staff is due to the X-ray scatter that is produced
when the X-ray beam interacts with the tissues of the patient. Thus, the higher the dose to
the skin of the patient the higher the amount of scattered radiation produced. From this,
patient and staff exposure are interlinked. One should always remember that the patient is
the main source of exposure of the staff. Therefore optimizing the patient dose will improve
the staff radiation protection.

4.1 Patient
Patient dose can be quite high but deterministic effects should never be a surprise to the
operator. Avoiding as much as possible the appearance of deterministic effects (short term
radiological complications) the operator should also minimize the stochastic effects (long
term radiological complications).

4.1.1 Patient positioning


The first thing to know is: where the source of radiation and the image detector are. The
patient needs to be as far as possible from the source of radiation (X-ray tube). The patient
needs to be as close as possible to the image detector.

4.1.2 X-ray tube orientation


The number of X-ray is reduced by a factor of two every 2 to 3 cm of tissue. Nevertheless,
the number of photons required to obtain an image remains constant. Thus, the thicker is
the patient the higher is the skin rate. Skin dose rate might be also very high when using
oblique incidence and one should try to restrict as much as possible incidences where the
path of the X-ray in the patient are long.

4.1.3 Image quality level


Fluoroscopy units provide the user with several image quality levels during fluoroscopy
and cine runs. There is no free lunch, the better the image quality the higher the patient
exposure. The cardiologist should be aware that he/she has control over the image quality
level and he/she should avoid any waste of radiation. The temporal resolution (frame rate)
should also be chosen according the requirement of the procedure.

4.1.4 Image magnification


Geometric magnification should never be used (see §4.1.1). Electronic or digital
magnification requires in general an increase of the skin dose rate. Thus, magnification
should be used with parsimony.

4.1.5 Introduction of an “intervention” level


During long procedures the threshold of the deterministic effects might be reached. Since
the severity of the lesion is dose dependent one should define a DAP level (for example 100
Gy.cm2) after which a particular care on patient exposure is exercised. One could try for
example to change incidences to distribute the dose. A DAP threshold where the patient
should be recalled to check for skin lesion should also be defined (for example 500 Gy.cm2).
374 Advances in the Diagnosis of Coronary Atherosclerosis

4.1.6 Patient dose record


The dose indicators (DAP and skin accumulated dose) of the procedure should be recorded
in the file of the patient. Moreover dose survey should be organised and the results should
be compared with published value to estimated one’s practice.

4.2 Personnel
The X-ray beam enters the patient and interacts with the tissue. Some radiation will be
absorbed in the patient and thus deposit energy to that patient. The transmitted radiation
will impinge the image amplifier to produce images. Unfortunately, the X-ray interaction
within the patient will also create scattered radiation emitted all around the patient which
will expose the staff to ionizing radiations.
The dose the staff might receive when being at one meter from the patient is approximately
0.1% of the skin dose delivered to that patient when the size of the X-ray beam is large
(typically 400 cm2). For example if the skin dose rate is equal to 40 mGy.min-1, ambient dose
rate at one meter from the patient can be estimated to 2.5 mSv.h-1 (natural background of
0.0001-0.0002 mSv.h-1). During cine runs the skin dose rate can reach 1 Gy.min-1 which
implies a higher dose rate to the staff during this type of acquisition. The ratio between
patient skin dose and staff dose is dependent on the size of the X-ray beam. The larger the
field size the higher the ratio. It is thus important to always reduce the size of the X-ray
beam to its strict minimum. Expressed in terms of DAP one can take 0.3% of the DAP (DAP
expressed in Gy.cm2 and E in mSv). Thus when a cardiologist has delivered a DAP of 60
Gy.cm2 he might have received over his/her lead apron a dose of 0.2 mSv.

4.2.1 Shielding
Shielding allows the staff to be isolated from scattered radiation produced by the X-ray
interaction in the patient. Barriers used for shielding may be fixed to the unit, movable or
worn by individuals. The amount of radiation attenuated by a material depends on the
elemental composition of the material, its thickness and the energy of the radiation passing
through it. As an example, the transmission through 0.5 mm of lead is 3.2% at 100kV and
0.36% at 70 kV (Yaffe & Mawdsley, 1991).
To reduce the operator exposure, one should begin by creating a sort of booth with
shielding devices around him or herself. To do so, an articulated ceiling leaded screen
lengthened by a leaded flap beside the table, or a mobile barrier as the one shown in Figure
1, together with a longitudinal protection adjacent to the table can be used (Kuon et al.
2002). Each shield should be at least 0.5 mm lead equivalent. The operator has then to
complete his or her protection by wearing a lead apron, which should be adjusted to his or
her size. Concerning the lead equivalent content, most of the aprons are either 0.5 or 0.35
mm lead equivalent. The use of a 0.35 mm instead of a 0.5 mm lead equivalent reduces
weight by about 30%, but increases the transmission of scattered radiation of almost a factor
of 2 at 90 kV. However, Marshall in an experimental study showed that at 90 kV, wearing a
0.35 mm lead equivalent apron with an additional 0.35 mm lead equivalent thyroid shield
resulted in a factor 2 greater reduction in effective dose than can be achieved by wearing a
0.55 mm lead equivalent apron alone (Marshall & Faulkner, 1993). Some aprons are made of
composite materials which offers equivalent shielding properties but which are significantly
less heavy than aprons containing lead as shield material (Yaffe & Mawdsley, 1991).
Figure 1 presents one way the operator can insulate himself from the scatter produced by
the patient when the examination is performed in the chest area with a femoral access. One
Protection of the Patient and the Staff from Radiation
Exposure During Fluoroscopy-Guided Procedures in Cardiology 375

should always remember that it is at the surface where the X-ray beam enters the patient
that the scatter dose rate is at its highest level. One should shield himself from that
particular spot.
Figure 2 summarizes the efficiency of the shielding devices presented in Figure 1 for various
tube-image amplifier orientations. It can be seen that an operator can significantly decrease
his or her exposure when a shielding device is placed in such a way that the operator is as
much as possible shielded from scattered radiation (Kuon et al., 2002). Unfortunately this is
not always possible, as in the case of biliary drainage for example. In such a case, staff
exposure is mainly controlled by the optimisation of patient exposure, the distance from the
scatter source and personal shielding (Williams, 1997).

Fig. 1. Example of operator shielding with various devices (1 leaded glass, 2 and 3 leaded
curtains, 4 leaded wall; 1 and 4 being on a mobile barrier)
To improve protection against cataract (which is a deterministic effect of ionising radiation),
a 0.5 mm lead equivalent eyeglass should be used to reduce exposure to the lens by 10% to
65% (Nicholson, 1995). Finally, gloves made of composite material and offering the same
tactile perception than regular chirurgical ones are available. Unfortunately, they offer a
limited protection since they usually reduce dose by a factor less than 2 (Vaño & Guibelalde,
1997; Balter, 2001).

4.2.2 Effect of the examination geometry


Backscattered radiation is the radiation that is scattered back from the surface at the beam
entrance. It is of high intensity because the entrance surface of the primary beam into the
patient has not been attenuated. Thus, ambient dose rate will always be significantly higher
when the operator is close to the x-ray tube, than when it is at the image amplifier side.
Shielding of the operator should be performed considering these parameters (see Figure 3)
(Brateman, 1999).
376 Advances in the Diagnosis of Coronary Atherosclerosis

Protection
Sv/h device
2000
without
1000
+
200
ambient dose rate

200
device (1+2)
100

20 +

10
10 device (3+4)
personal
0.1
shielding

PA - RAO LAO 90º LAO 60º ± 20º


Tube - Image Amplifier incidence

Fig. 2. Mean ambient dose rate to the operator (Sv.h-1) for different tube-image amplifier
orientations. The dose rate corresponds to an examination in the cardiac area of a Rando
phantom, representative of a 70 kg standard patient, the operator being at about 1 m from
the central beam impinging the phantom (situation presented in Figure 1). Without any
radiation protection device, the dose rate is in the range of 2 to 0.2 mSv.h-1. As expected, the
LAO angulations are the ones which potentially deliver the highest dose to the operator.
The addition of the protection devices 1 and 2 allows a reduction of the ambient dose rate at
operator’s level by a factor of 10. A further reduction is obtained by completing the
shielding with devices 3 and 4. Finally, the use of personal shielding devices (i.e. lead apron,
thyroid shield and leaded glasses) reduces the ambient dose rate to about 0.1 Sv.h-1
(adapted from Kuon et al., 2002).

X Ray tube
Image Amplifier

Primary
Radiation Scattered
Radiation

Primary
Radiation

Scattered
Image Amplifier Radiation

X Ray tube

Fig. 3. The schematics show the influence of the tube-image amplifier orientation on the
operator’s exposure. When the tube is below the table, exposure to the legs and the lower
part of the abdomen is the highest. When the tube is above the table, exposure to the face,
neck and chest is the highest.
Protection of the Patient and the Staff from Radiation
Exposure During Fluoroscopy-Guided Procedures in Cardiology 377

4.2.3 Inverse square law


The exposure rate from a point source of radiation decreases as the distance from the source
squared. This inverse square law is the results of the geometric relationship between the
surface area, A, and the radius, r, of a sphere: A = 4 п r2. As an example, for a field size of
the primary beam impinging the patient of about 100 cm2, the ambient dose rate at 90° to the
incident beam decreases from 1.2 mGy.h-1 to 0.3 mGy.h-1 when moving from 50 cm to 1m
from the patient (i.e. a dose rate reduction as a function of 1/r2) (Bushberg et al., 2002).
Unfortunately, when the size of the field impinging the patient is large, the dose reduction is
slightly less efficient since dose rate reduction is no more as a function of r-2 but only as a
function of r-1. Nevertheless, backing away from the primary beam is a very efficient way to
reduce staff exposure.

5. Conclusion
Several recently published studies showed that there is a high potential to reduce radiation
doses to the patient and subsequently to enhance radiation safety for the staff. The aim of
this chapter was to introduce the effects of ionizing radiation and the radiological risks, and
to present various methods that may be used to reduce patient and personnel exposure
during fluoroscopy-guided procedures in cardiology, such as the reduction of patient
exposure, the increase of the distance from the source of scatter, and the shielding.
Every effort needs to be made in order to reduce patient and staff exposure as much as
possible, not for legal purposes but simply for workers health. This applies in particular in
the field of interventional radiology where exposure to the staff is potentially very high.
Dose reduction to the staff can be reduced by the optimisation of patient exposure. Thus the
introduction of the diagnostic reference levels (DRL) will certainly improve the control of
staff exposure. Moreover, the application of simple rules, such as the ones mentioned in this
contribution, allow radiologists to use fluoroscopy units minimising their exposure as much
as possible. The respect of these rules allow also to improve the protection of other medical
staff present near the patient and who are less familiar with radiation protection.

6. References
Einstein, A.J.; Moser, K.W., Thompson R.C., Cerqueira M.D., Henzlova M.J. (2007).
Radiation dose to patients from cardiac diagnostic imaging. Circulation, 116:1290-
1305
Samara, E.; Aroua, A., Vader, J.-P., Trueb, Ph., Bochud, F., Verdun, F.R. (2011). Exposure of
the Swiss Population by Radiodiagnostics: 2008 Review. Health Physics, submitted
Suzuki, S.; Furui, S., Isshiki, T. (2008). Patients' skin dose during percutaneous coronary
intervention for chronic total occlusion. Catheter Cardiovasc Interv, 71:160-164
Jacobi, W.; (1975). The concept of effective dose – a proposal for the combination of organ
doses Radiat Environ Biophys, 12:101-109
ICRP 26 (1977). International Commission on Radiological Protection: The recommendations
of the international commission on radiological protection. ICRP Publication 26,
Elsevier
ICRP 90 (1990). International Commission on Radiological Protection: The recommendations
of the international commission on radiological protection. ICRP Publication 90,
Elsevier
378 Advances in the Diagnosis of Coronary Atherosclerosis

ICRP 103 (1007). International Commission on Radiological Protection: The


recommendations of the international commission on radiological protection. ICRP
Publication 103, Elsevier
ICRU 47 (1992). International Commission on Radiation Units and Measurements. ICRU
Report 47, ICRU, Bethesda, Maryland, USA (1992).
Yaffe, M.J. & Mawdsley, G.E. (1991). Composite materials for x-ray protection. Health Phys
60:661-664
Kuon, E.; Schmitt, M. & Dahm, J.B. (2002). Significant reduction of radiation exposure to
operator and staff during cardiac interventions by analysis of radiation leakage and
improved lead shielding. Am J Cardiol 89: 44-49
Marshall, N.W. & Faulkner, K. (1993). Optimization of personnel shielding in interventional
radiology. Proceedings of the Radiation Protection Committee of the BIR (British
Institute of Radiology) and of the Commission of the European Communities
(CEC). London
Williams; J.R.; (1997). The interdependence of staff and patient doses in interventional
radiology. Br J radiol 70: 498-503
Vaño, E. & Guibelalde, E. (1997). Proceedings of the European radiation protection
education and training ERPET course – Radiation protection in interventional
radiology. Madrid 12-14 May 1997, European Commission DG XII, Brussels
Balter, S.; (2001). Stray radiation in the cardiac catheterisation laboratory. Radiat Prot Dosim
94(1-2): 183-188
Brateman, L.; (1999) The AAPM/RSNA Physics tutorial for residents – Radiation safety
considerations for diagnostic radiology personnel. Radiographics 19:1037-1055
Bushberg, J.T.; Seibert, J.A., Leidholdt, E.M., Boone, J.M. (2002). The essential physics of
medical imaging. Lippincott Williams & Wilkins, Philadelphia

You might also like