10.05.2015 Views

Mar-Apr 2013 - Indian Heart Journal

Mar-Apr 2013 - Indian Heart Journal

Mar-Apr 2013 - Indian Heart Journal

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

2 nd World Summit<br />

on Echocardiography<br />

October 25-27, <strong>2013</strong> | new delhi, india<br />

leela Kempinski Gurgaon Hotel<br />

REGISTRATION NOW OPEN | www.wsecho.org<br />

An EduCAtiOnAl COllAbOrAtiOn by:<br />

• American Society of Echocardiography • European Association of Cardiovascular Imaging,<br />

• <strong>Indian</strong> Academy of Echocardiography<br />

a branch of the European Society of Cardiology<br />

• Echocardiography Association of the<br />

Interamerican Cardiology Society<br />

• Japanese Society of Echocardiography<br />

• Korean Society of Echocardiography<br />

• Canadian Society of Echocardiography<br />

• Chinese Society of Echocardiography<br />

Abstract Submissions Accepted <strong>Mar</strong>ch 1 – May 31, <strong>2013</strong><br />

Partho P. Sengupta, MBBS, MD, DM, FASE<br />

International Chair<br />

Jagdish C. Mohan, md, dm<br />

Program Chair


indian heart journal 65 (<strong>2013</strong>) 152e157<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original article<br />

Comparative assessment of platelet Gp IIb/IIIa receptor<br />

occupancy ratio with Eptifibatide/Tirofiban in patients<br />

presenting with ACS and undergoing PCI<br />

Aniket Puri a, *, A. Bansal b , V.S. Narain c , R. Sethi d , S.K. Dwivedi c , V.K. Puri e , R.K. Saran f<br />

a Associate Professor, Department of Cardiology, CSM Medical University (Erst. King George Medical University), B-58 Sector A, Mahanagar,<br />

Lucknow, India<br />

b Senior Resident, Department of Cardiology, CSM Medical University (Erst. King George Medical University), Lucknow, India<br />

c Professor, Department of Cardiology, CSM Medical University (Erst. King George Medical University), Lucknow, India<br />

d Assistant Professor, Department of Cardiology, CSM Medical University (Erst. King George Medical University), Lucknow, India<br />

e Former Professor, Department of Cardiology, CSM Medical University (Erst. King George Medical University), Lucknow, India<br />

f Professor and Head, Department of Cardiology, CSM Medical University (Erst. King George Medical University), Lucknow, India<br />

article info<br />

Article history:<br />

Received 28 February 2012<br />

Received in revised form<br />

30 May 2012<br />

Accepted 12 August 2012<br />

Available online 27 August 2012<br />

Keywords:<br />

Glycoprotein IIb/IIIa (Gp IIb / IIIa )<br />

antagonists<br />

Acute coronary syndrome<br />

Percutaneous coronary intervention<br />

abstract<br />

Background: The level of platelet inhibition by a Glycoprotein IIb/IIIa (Gp IIb/IIIa ) antagonist<br />

therapy necessary to minimize thrombotic complications in patients undergoing percutaneous<br />

coronary intervention (PCI) is a subject of debate. The degree of platelet inhibition<br />

obtained 10 min after start of Gp IIb/IIIa antagonist therapy predicts adverse events after PCI.<br />

The aim of this study was to look at platelet inhibition and to compare platelet Gp IIb/IIIa<br />

receptors occupancy ratio (GpRO) with Eptifibatide and Tirofiban using various dose regimens<br />

and correlate with 30-day clinical outcomes in patients presenting with high-risk<br />

acute coronary syndromes (ACS) and undergoing PCI.<br />

Methods: The patients were divided into four sub groups: (1) Eptifibatide two intracoronary<br />

bolus (180 mg/kg) alone (E B ); or (2) two intravenous bolus (180 mg/kg) followed by infusion at<br />

2 mg/kg/min for 24 h (E B þ Inf ); and (3) Tirofiban standard bolus dose (0.4 mg/kg) over 30 min<br />

followed by infusion at 0.1 mg/kg/min (T Std ); or (4) at ADVANCE dose bolus (25 mg/kg) over<br />

3 min, followed by infusion at 0.1 mg/kg/min (T Adv ). Number of Gp IIb/IIIa receptors was<br />

assessed by flow cytometry at baseline and 10 min after the bolus and percentage of free<br />

receptors was determined to calculate the GpRO. Patients were followed for 30 days for any<br />

major adverse cardiac events (MACE).<br />

Results: 200 consecutive patients (including 74% with ST-elevation ACS) were enrolled.<br />

GpRO in groups E B (n ¼ 48) and E B þ Inf (n ¼ 44) were 62.7% 27.2% and 61.4% 6.1%<br />

respectively while in the groups T Std (n ¼ 96) and T Adv (n ¼ 12) groups were 35.1% 17.74%<br />

and 68.8% 27.3% respectively. The GpRO was similar in E B ,E B þ Inf and T Adv groups and<br />

was significantly higher than T Std group (p < 0.0001). The 30-day MACE rates in E B (4.2%),<br />

E B þ Inf (4.5%) and T Adv (4.2%) were significantly lower than T Std group (12.5%) (p < 0.01).<br />

* Corresponding author.<br />

E-mail address: aniketpuri@hotmail.com (A. Puri).<br />

0019-4832/$ e see front matter Copyright ª 2012, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.2012.08.007


indian heart journal 65 (<strong>2013</strong>) 152e157 153<br />

Conclusions: Standard dose Tirofiban results in significantly lower rates of Gp IIb/IIIa receptor<br />

occupancy ratio and this correlated with higher incidence of 30-day MACE in high-risk ACS<br />

patients undergoing PCI.<br />

Copyright ª 2012, Cardiological Society of India. All rights reserved.<br />

1. Introduction<br />

The level of platelet inhibition induced by a Glycoprotein IIb/IIIa<br />

(Gp IIb/IIIa ) receptor antagonist therapy necessary to minimize<br />

thrombotic complications in patients undergoing percutaneous<br />

coronary intervention (PCI) has been the subject of various<br />

studies. Steinhubl et al, in the GOLD study, showed that the<br />

degree of platelet function inhibition obtained 10 min after the<br />

start of Gp IIb/IIIa receptor antagonist therapy is an independent<br />

predictor of the risk of major adverse cardiac events after PCl. 1<br />

It has been shown that the extent of inhibition of platelet aggregation<br />

depends on various factors like, (1) the clinical presentation,<br />

(2) the type of antiplatelet agents used and (3) the dose of the<br />

antiplatelet agent. However, considerable inter patient variability<br />

in the platelet inhibitory response has been observed in various<br />

studies. The effort to achieve and sustain optimal receptor<br />

blockade had led to various adjustments in the weight-based<br />

dosing strategies of the different Gp IIb/IIIa receptor antagonist<br />

drugs namely Abciximab, Eptifibatide and Tirofiban. Initial dose<br />

finding trials in humans attempted to identify the appropriate<br />

dosing regimen necessary to achieve and maintain >80% Gp IIb/IIIa<br />

receptor blockade, or achieve 80%<br />

of the receptors completely abolished ADP induced platelet<br />

aggregation, suggesting a steep doseeresponse curve. 2,3<br />

Quantitative flow cytometry allows platelet membrane<br />

receptor measurement in whole blood samples and Glycoprotein<br />

IIb/IIIa receptor occupancy (GpRO) ratio can be<br />

measured either directly or indirectly. <strong>Mar</strong>kers of platelet<br />

activation have been studied with flow cytometry to predict<br />

an increased risk of ischemic events after PCI. It has been<br />

shown that higher levels of platelet GpRO with Eptifibatide<br />

have been associated with improved myocardial perfusion<br />

among patients with ST-elevation myocardial infarction. It<br />

has also been shown that intracoronary administration of<br />

Eptifibatide results in a high local concentration, which in turn<br />

may lead to increased levels of platelet GpRO, cause more<br />

destabilization of platelet aggregates, and promote thrombus<br />

disaggregation, thereby improving myocardial perfusion. 4,5<br />

The aim of this study was to compare the platelet Gp IIb/IIIa<br />

receptors occupancy ratio with Eptifibatide and Tirofiban<br />

using flow cytometry and to correlate with 30-day clinical<br />

outcomes in patients undergoing PCI for established clinical<br />

indications also being treated with Gp IIb/IIIa receptor antagonist<br />

therapy.<br />

receiving a Gp IIb/IIIa receptor antagonist therapy in the form of<br />

either Eptifibatide or Tirofiban based on the treating clinician’s<br />

discretion. The index diagnosis of ACS included unstable angina,<br />

noneST-elevation myocardial infarction (NSTEMI), or STelevation<br />

myocardial infarction (STEMI). All STEMI patients<br />

included were treated as per best prevailing indications including<br />

with thrombolytic therapy and decision for PCI was based on the<br />

physician’s discretion, which in turn was based on current best<br />

clinical practice with most patients having routine angiography<br />

within 24 h and proceeding for PCI if needed. The exclusion<br />

criteria were multivessel PCI at the time of the index procedure;<br />

severe coronary calcification; unprotected left main stenosis<br />

>50%; target lesion in a saphenous vein graft; previous stenting<br />

at the target lesion; or a contraindication to Gp IIb/IIIa receptor<br />

antagonist therapy. The age, sex, detailed clinical history<br />

including that of diabetes mellitus, smoking, hypertension,<br />

family history of coronary artery disease (CAD), hospitalization<br />

for CAD were noted. At admission a thorough clinical examination<br />

including measuring the body weight (Wt), pulse rate (PR),<br />

systolic blood pressure (SBP), determination of Killip class (KC),<br />

and an electrocardiogram (ECG) was done. In addition to this,<br />

Troponin T, lipid profile, platelet count before and after PCI, CPK-<br />

MB the day following PCI were also recorded. 2D Echocardiogram<br />

was done for determination of left ventricular ejection fraction<br />

(EF) using Simpson’s method, along with LV end diastolic and end<br />

systolic volume noted. Informed consent for participation in the<br />

study was obtained by all patients prior to enrollment.<br />

During PCI, all of the patients were treated as per protocol<br />

with at least 325 mg aspirin before the procedure. A loading dose<br />

of Clopidogrel 300 mg was administered prior to the PCI. A<br />

weight-adjusted heparin regimen was used to titrate and achieve<br />

a minimum activated clotting time of 250 s. Guide wires and<br />

approved stents (drug-eluting or bare metal) were used according<br />

to standard techniques and based on the performing physician’s<br />

discretion. Stent was deployed after completion of the<br />

bolus dose of Gp IIb/IIIa receptor antagonist therapy, and choice of<br />

drug [Eptifibatide (Group E) or Tirofiban (Group T)] was left to the<br />

performing physicians discretion and hence the study was not<br />

randomized or blinded to one treatment or another upon<br />

enrollment. Each group was further divided into two subgroups<br />

each based on the dose regimen used viz (1) Eptifibatide two<br />

intracoronary bolus (180 mg/kg) alone (E B ); or (2) two intravenous<br />

bolus (180 mg/kg) followed by infusion at 2 mg/kg/min (E B þ Inf ); and<br />

(3) Tirofiban standard dose (T Std )(0.4mg/kg bolus over 30 min)<br />

followed by 0.1 mg/kg/min infusion; or (4) at ADVANCE dose (T Adv )<br />

(25 mg/kg bolus over 3 min), followed by infusion at 0.1 mg/kg/min<br />

based on the dose used in the ADVANCE trial. 6<br />

2. Methods<br />

We included all consecutive patients admitted to our hospital<br />

undergoing PCI for an acute coronary syndrome (ACS) and<br />

3. Platelet aggregometry study<br />

Ten ml blood samples were taken in 3.8% sodium citrate vials,<br />

at baseline from the catheter prior to engaging the coronary


154<br />

indian heart journal 65 (<strong>2013</strong>) 152e157<br />

ostia and 10 min after both boluses of Eptifibatide; or in case of<br />

Tirofiban 10 min after starting the infusion. Samples were<br />

analyzed at an ISO 9001:2000 and NABL certified laboratory by<br />

flow cytometry using platelet Gp IIb/IIIa Occupancy kit (PLT<br />

VASP/P2Y12, Biocytex, <strong>Mar</strong>seille, France). At baseline, the<br />

numbers of Gp IIb/IIIa receptor/cell were calculated and the<br />

percentage of free receptors was determined. With the 2 nd<br />

sample, again percentage of free and bound receptors was<br />

calculated and subsequently Gp IIb/IIIa receptor occupancy<br />

(GpRO) ratio was determined. The flow cytometric method is<br />

based on binding of two monoclonal antibodies to platelet<br />

Gp IIb/IIIa viz Mab1 and Mab2. Monoclonal antibody 1 (Mab 1,<br />

Clone L4P18) is a murine monoclonal antibody to Gp IIb/IIIa<br />

complex and the monoclonal antibody 2 (Mab 2, Clone 4F8) is<br />

a murine monoclonal antibody to beta 3 subunits. Binding of<br />

these antibodies is used in a flow cytometric assay to directly<br />

quantify occupied and unoccupied Glycoprotein IIb/IIIa<br />

receptors. Eptifibatide binds with Mab 2 in a dose-dependent<br />

manner and has little effect on Mab 1 binding while Tirofiban<br />

binds with both Mab 1 and Mab 2. The total number of platelet<br />

Gp IIb/IIIa receptors and number of free Gp IIb/IIIa receptors are<br />

determined by converting the fluorescence intensity into the<br />

corresponding numbers of sites per platelet base on a calibrated<br />

bead standard curve. The final GpRO results for each<br />

patient were blinded till the end of study period.<br />

Major adverse clinical event (MACE) end points of recurrent<br />

ischemia, reinfarction, target vessel revascularization, worsening<br />

heart failure, repeat hospitalization for cardiovascular<br />

(CV) causes and CV Death during these 30 days were recorded.<br />

Worsening heart failure was defined as a worsening in Killip’s<br />

class of more than one grade. Recurrent ischemia was defined<br />

as those who had a worsening of NYHA functional class of<br />

more than one grade during the 30 days and required a step<br />

up of anti-anginal therapy. Those who could not be stabilized<br />

on medical therapy and required hospitalization either for<br />

recurrent ischemia or worsening heart failure, were included<br />

in the group of repeat hospitalizations. The entire study was<br />

cleared by the ethics committee of the university and conformed<br />

to the guidelines set for good clinical practice.<br />

3.1. Statistical analysis<br />

SPSS 11.5 software was used for analysis of the data obtained.<br />

The student t test, Fisher’s exact test and Chi square test were<br />

used to test the significance between the study groups. Risk<br />

analysis was carried out by calculating the odds ratio (OR) and<br />

95% confidence interval (CI).<br />

4. Results<br />

Two hundred consecutive patients were enrolled in the study.<br />

The mean age of the patients was 53 years. The index event<br />

and indication of PCl was ST-elevation myocardial infarction<br />

(STEMI) in 74% (38% with anterior wall myocardial infarction<br />

(AWMI), 36% with inferior wall myocardial infarction (IWMI));<br />

noneST-elevation myocardial infarction (NSTEMI) in 22%, and<br />

unstable angina 4%; in the STEMI group 10% patients had<br />

primary PCI and 6% had rescue PCI following failed<br />

thrombolysis.<br />

Among the cohort 20% patients were diabetic, 32% were<br />

hypertensive, 32% had dyslipidemia, 50% were smokers and<br />

38% patients had 2 risk factors. The mean level of troponin<br />

T elevation on admission was 2.25 ng/ml. Baseline characteristics<br />

of the patient’s are shown in Table 1.<br />

Based on the drugs used the patients were divided into two<br />

groups i.e. the Eptifibatide group (Group E) and the Tirofiban<br />

group (Group T) and further subdivided into four subgroups<br />

i.e., (1) Eptifibatide two intracoronary bolus alone (Group E B )<br />

which included 48 patients; (2) Eptifibatide intravenous bolus<br />

followed by infusion (Group E B þ Inf ) which included 44<br />

patients; (3) Tirofiban standard dose group (T Std ) which<br />

included 96 patients; and (4) Tirofiban ADVANCE dose (Group<br />

T Adv ) which included 12 patients.<br />

4.1. Gp IIb/IIIa occupancy ratio<br />

The average numbers of Gp IIb/IIIa receptors/cell at baseline was<br />

73,379 12,426 in the entire cohort. Based on patients<br />

risk factors in the diabetic subgroup the average number of<br />

Gp IIb/IIIa receptors/cell was 75,200 10,115; in hypertensive<br />

patients it was 71,437 11,225; in smokers it was<br />

72,920 10,246; and in the dyslipidemia group it was<br />

76,687 10,356. At baseline 6.12% 8.62% Gp IIb/IIIa receptors<br />

were occupied, which could be attributed to receptor binding<br />

with fibrinogen and von Willebrand factor. The GpRO ratio<br />

above baseline in Group E was 70.25% 19.11% while in the<br />

Group T was 38.20% 20.74% (p < 0.001).<br />

The absolute GpRO was significantly higher with Group E<br />

62.1% 17.1% as compared to Group T 38.8% 18.8%<br />

(p < 0.001). Across all patient risk groups Eptifibatide achieved<br />

significantly higher GpRO as compared with Tirofiban viz, in<br />

diabetic patients 61.16% 26% v/s 38.53% 22.23% (p < 0.05);<br />

in hypertensive patients 56.82% 21.56% v/s 37.57% 22.89%<br />

Table 1 e Baseline characteristics of patients (n [ 200).<br />

Patient<br />

demographics<br />

Eptifibatide<br />

(n ¼ 92)<br />

Tirofiban<br />

(n ¼ 108)<br />

Mean age 53 52<br />

Male 76 96<br />

Female 16 12<br />

Diabetes mellitus 24 16<br />

Hypertension 40 24<br />

Dyslipidemia 32 32<br />

Smoker 40 60<br />

Indications of PCI<br />

USA 0 8<br />

NSTEMI 12 32<br />

STEMI 80 68<br />

AWMI 56 20<br />

IWMI 24 48<br />

Urgency of PCI<br />

Elective PCI 72 96<br />

Primary PCI 12 8<br />

Rescue PCI 8 4<br />

PCI ¼ Percutaneous Coronary Intervention; USA ¼ unstable angina;<br />

NSTEMI ¼ noneST-elevation myocardial infarction; STEMI ¼ STelevation<br />

myocardial infarction; AWMI ¼ Anterior Wall myocardial<br />

infarction; IWMI ¼ Inferior Wall myocardial infarction.


indian heart journal 65 (<strong>2013</strong>) 152e157 155<br />

(p < 0.05); in dyslipidemia patients 67.6% 13.49% v/s<br />

30.85% 17.31% (p < 0.05); while there was a non-significant<br />

increase in GpRO with Eptifibatide in smokers as compared<br />

to Tirofiban 53.31% 23.69% v/s 43.72% 18.02% (Table 2). In<br />

the presence of visible thrombus the GpRO was significantly<br />

lower as compared to without visible thrombus in Group E<br />

with visible thrombus 56.24% 22.96% v/s 75.74% 13.24%<br />

with no visible thrombus (p < 0.01); and in Group T with visible<br />

thrombus 29.60% 18.7% v/s 41.20% 18.84% with no visible<br />

thrombus (p < 0.01).<br />

The GpRO based on the index event of the patient was<br />

51.12% 23.75% in the STEMI patients; 38.61% 22.74% in the<br />

NSTEMI patients and 63.23% 18.7% in the unstable angina<br />

patients. Interestingly, there was a trend of increased GpRO in<br />

patients having platelet count greater than 250,000. The GpRO<br />

achieved according to weight showed significantly higher<br />

GpRO in the patients having less than 60 kg body weight in<br />

Group E 67.77% 27.74% v/s 59.78% 19.74% (p < 0.05) and in<br />

Group T 46.77% 20.13% v/s 31.56% 17.16% (p < 0.05).<br />

In the subgroup E B absolute GpRO was 62.7% 27.2% while<br />

in the subgroup E B þ Inf absolute GpRO was 61.4% 6.1%;<br />

whereas in subgroup T Std absolute GpRO was 35.1% 17.74%<br />

and in subgroup T Adv was 68.8% 27.3%. Therefore the<br />

absolute GpRO was similar in subgroups E B ,E B þ Inf and T Adv<br />

groups which was significantly higher than T Std group<br />

(p < 0.001). The percentage of patients achieving >80% GpRO<br />

was only seen with 13% in the E B þ Inf group, 9% in the E B group,<br />

8% in the T Adv group and in none of the patients in the T Std<br />

group. There was no difference in any bleeding side effects<br />

across all four subgroups following the procedure.<br />

The incidence of 30-day MACE rates in Group E was 4.3%<br />

(subgroup E B 4.2% and subgroup E B þ Inf 4.5%), while in Group T<br />

was 13% (subgroup T Adv 8.3% and subgroup T Std 12.5%). There<br />

was a trend for MACE to be lower with Eptifibatide as<br />

compared to Tirofiban although this did not reach statistical<br />

significance (p ¼ 0.06). MACE rates were lower in E B ,E B þ Inf and<br />

T Adv subgroups than T Std subgroup, however this did not<br />

reach statistical significance (Table 3).<br />

5. Discussion<br />

Glycoprotein IIb/IIIa receptor antagonists have been shown<br />

to therapeutically down regulate platelet function to prevent<br />

the thrombotic complications associated with coronary<br />

artery disease. 7 However, several individual studies, in<br />

specific patient populations or with suboptimal dosing regimens,<br />

have found a lack of effect in preventing thrombotic<br />

complications. 8 These results raise the question as to<br />

whether a specific dose of Gp IIb/IIIa receptor antagonist,<br />

adjusted only by patient weight, can provide the same level<br />

of platelet inhibition across all clinical syndromes and across<br />

all individuals.<br />

The GOLD multicenter study, 1 showed that patients who<br />

did not achieve >95% inhibition of Gp IIb/IIIa receptors after<br />

bolus of Gp IIb/IIIa receptor antagonist therapy experienced<br />

a significantly higher incidence of MACE rates (14.4% v/s 6.4%,<br />

p ¼ 0.006). Patients whose platelet function was 70% inhibition of platelet function<br />

(p ¼ 0.009) and on multivariate analysis it was found that<br />

platelet function inhibition >95% at 10 min after the start of<br />

therapy was associated with a significant decrease in the<br />

incidence of MACE. However, in our study only 7% of patients<br />

in the entire cohort achieved >80% GpRO (13% in the E B þ Inf<br />

group, 9% in the E B group, 8% in the T Adv group and in none of<br />

the patients in the T Std group) which may indicate suboptimal<br />

drug effect or variation in pharmacological response in our<br />

population.<br />

It has been suggested that the disaggregation of platelet<br />

thrombi may be a mechanism for the clinical benefits. Eptifibatide<br />

has demonstrated to reduce the occurrence of myonecrosis<br />

with PCI in acute coronary syndromes, a clinical<br />

scenario associated with platelet rich thrombus development.<br />

In an analysis of the Enhanced Suppression of the Platelet IIb/<br />

IIIa Receptor With Integrilin Therapy (ESPRIT) trial, patients<br />

were then stratified into high- and low-risk groups in which<br />

thrombotic complications with the PCI were more likely<br />

during revascularization. High-risk characteristics included<br />

age >75 years, diabetes, ST-segment elevation within 7 days,<br />

or unstable angina within 48 h. The high-risk group demonstrated<br />

a reduction in the combined end point of death or<br />

myocardial infarction at both 30 days (6.2% v/s 12.4%;<br />

p < 0.001) and 12 months (8.0% v/s 15.9%; p < 0.001). 9 The lowrisk<br />

group demonstrated a trend toward benefit but did not<br />

achieve a statistically significant improvement with Eptifibatide<br />

thereby suggesting that higher risk groups benefit more.<br />

In our patient population, the GpRO achieved based on the<br />

Table 2 e Glycoprotein IIb/IIIa receptor occupancy ratio and 30-day major adverse clinical event across Eptifibatide group<br />

(Group E) and Tirofiban group (Group T).<br />

Results Group E, n ¼ 92 Group T, n ¼ 108 T value p Value<br />

GpRO above baseline 70.3% 19.1% 38.2% 20.7% 4.3


156<br />

indian heart journal 65 (<strong>2013</strong>) 152e157<br />

Table 3 e Subgroups analysis of Glycoprotein IIb/IIIa receptor occupancy ratio and 30-day major adverse clinical event,<br />

across the four subgroups.<br />

Results Group E B , n ¼ 48 Group E B þ Inf , n ¼ 44 Group T Adv , n ¼ 12 Group T Std , n ¼ 96 p Value<br />

Absolute GpRO 62.7% 27.2% 61.4% 6.1% 68.8% 27.3% 35.1% 17.7% 80% 9% 13% 4% 0%


indian heart journal 65 (<strong>2013</strong>) 152e157 157<br />

10. Deibele AJ, Jennings LK, Tcheng JE, Neva C, Earhart AD,<br />

Gibson CM. Intracoronary eptifibatide bolus administration<br />

during percutaneous coronary revascularization for acute<br />

coronary syndromes with evaluation of platelet glycoprotein<br />

IIb/IIIa receptor occupancy and platelet function: the<br />

intracoronary eptifibatide (ICE) trial. Circulation.<br />

2010;121:784e791.<br />

11. Fischell TA. “Bolus-only” glycoprotein IIb/IIIa inhibitor use for<br />

elective percutaneous coronary intervention: maybe less is<br />

more? Am <strong>Heart</strong> J. 2006;152:812e814.


indian heart journal 65 (<strong>2013</strong>) 191e193<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Case report<br />

Absent pulmonary valve syndrome with tetralogy of Fallot<br />

detected at an early gestational age of 27 weeks e A case<br />

report<br />

Ashok N. Bhupali a,b , Kiran B. Patankar c , Sayi Prasad d , Jeetendra K. Patil e ,<br />

Ajitey Tamhane f, *<br />

a Consultant Interventional Cardiologist and Head of Institute, Saraswati Hospital and Advanced Medical Care Center<br />

(Post Graduate Institute-DNB), Dasara Chowk, Kolhapur 416002, Maharashtra, India<br />

b Consultant Cardiologist, Apple Hospital and Research Institute, Diagnostic Center, Shahupuri, Kolhapur, Maharashtra, India<br />

c Director (Radiology), Apple Hospital and Research Institute, Diagnostic Center (Post Graduate Institute-DNB), Shahupuri,<br />

Kolhapur, Maharashtra, India<br />

d Consultant Physician and Intensivist, Saraswati Hospital and Advanced Medical Care Center (Post Graduate Institute-DNB),<br />

Dasara Chowk, Kolhapur 416002, Maharashtra, India<br />

e Head of Department of Advanced Imaging Radiology (CT & MRI), Apple Hospital and Research Institute Diagnostic Center<br />

(Post Graduate Institute-DNB), Shahupuri, Kolhapur, Maharashtra, India<br />

f Post Graduate Resident in Radiodiagnosis, Apple Hospital and Research Institute, Diagnostic Center (Post Graduate Institute-DNB),<br />

525, E, Vyapari Peth, Shahupuri, Kolhapur, Maharashtra, India<br />

article info<br />

Article history:<br />

Received 11 July 2012<br />

Accepted 23 October 2012<br />

Available online 29 October 2012<br />

Keywords:<br />

Absent pulmonary valve syndrome<br />

Tetralogy of Fallot<br />

Congenital heart Anomalies<br />

Fetal echocardiography<br />

abstract<br />

Objective: Absent pulmonary valve syndrome (APVS) is a rare congenital anomaly, usually<br />

seen in association with a ventricular septal defect. It has been reported to occur in 3e6% of<br />

cases of tetralogy of Fallot (TOF). In this case report we discuss a case of absent pulmonary<br />

valve syndrome with tetralogy of Fallot that was detected in utero by fetal echocardiography<br />

at 27 weeks of gestation.<br />

Case: A 20-year-old pregnant woman at 27 weeks of gestation referred to our Institute. She<br />

has no consanguineous history. We diagnosed the case as tetralogy of Fallot with absent<br />

pulmonary valves in fetal echocardiographic study.<br />

Conclusion: We conclude that when a paracardiac cystic, pulsatile lesion with dilated<br />

pulmonary arteries are seen in the fetus in utero then other features associated with the<br />

syndrome, such as TOF and the presence or absence of the ductus arteriosus should be<br />

looked for. In our case there was no ductus arteriosus.<br />

Copyright ª 2012, Cardiological Society of India. All rights reserved.<br />

1. Introduction<br />

Absent pulmonary valve syndrome (APVS) is a rare congenital<br />

anomaly, usually seen in association with a ventricular septal<br />

defect. It has been reported to occur in 3e6% of cases of<br />

tetralogy of Fallot (TOF). Absence of the pulmonary valve<br />

results in a dilated main pulmonary artery, which can be seen<br />

as a cystic, pulsatile, paracardiac lesion on antenatal USG.<br />

* Corresponding author. Tel.: þ91 231 2651207, þ91 9823265255 (mobile); fax: þ91 231 2654850.<br />

E-mail address: drtamhanes@hotmail.com (A. Tamhane).<br />

0019-4832/$ e see front matter Copyright ª 2012, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.2012.10.010


192<br />

indian heart journal 65 (<strong>2013</strong>) 191e193<br />

Such a lesion, though rare, can easily be detected. We report<br />

a case of this rare anomaly which was present in association<br />

with a TOF. The case was detected at 27 weeks of gestation.<br />

The prognosis depends on the respiratory complications. 1<br />

When APVS is associated with a ventricular septal defect,<br />

the physiologic and anatomic repercussions affect both<br />

ventricles, and cardiac performance can be critically<br />

impaired. 2 We report a case of APVS that was detected by USG<br />

at 27 weeks of gestation; As of now the fetus is grossly normal<br />

and growth is corresponding to gestational age.<br />

2. Case report<br />

A 20-year-old primigravida presented for routine prenatal USG<br />

scanning. There was a single fetus of about 27 weeks gestation.<br />

The fetus showed a pulsatile cystic lesion located near<br />

the heart. We therefore performed a detailed fetal echocardiography,<br />

which revealed. The superior and inferior vena<br />

cava drained normally into the right atrium. Both Atria were<br />

normal. Atrioventricular concordance was noted. The<br />

tricuspid and mitral valves were normal. The right ventricle<br />

(RV) was seen to open into the pulsatile cystic lesion, which<br />

was thus confirmed to be the dilated main pulmonary artery<br />

along with the right and left pulmonary arteries. The pulmonary<br />

annulus showed no pulmonary valve (p-valve) echoes<br />

[Fig. 1]. There was back-and-forth flow across the RV and the<br />

dilated pulmonary artery during systole and diastole of the RV<br />

[Fig. 2]. The aorta was seen arising from the left ventricle (LV)<br />

and there was 50% overriding of aorta. A large subaortic<br />

ventricular septal defect (VSD) was seen [Fig. 3]. On Color<br />

Doppler study pulmonary regurgitation was noted [Fig. 4].<br />

Other systems were unremarkable. Thus, a diagnosis of TOF<br />

with absent pulmonary valves was made.<br />

3. Discussion<br />

APVS is a complex syndrome comprising dysplasia/absence<br />

of pulmonary valvular leaflets, with resultant regurgitation<br />

Fig. 2 e An axial color Doppler image of the right<br />

ventricular outflow tract (arrows) shows the dilated<br />

pulmonary artery (PA) and the right ventricle (RV) with<br />

a mixed hue of colors suggesting to-and-fro random flow<br />

between the right ventricle and the pulmonary artery.<br />

and dilatation of the main and branch pulmonary arteries. 3<br />

The majority of these cases present with a VSD and features<br />

of TOF. The aneurysmal dilatation of the pulmonary artery<br />

often results in compression of the bronchial tree and<br />

esophagus, with consequent bronchomalacia and polyhydramnios.<br />

Volpe et al studied 21 fetuses with APVS for their<br />

associations and outcomes. Their study reveals an association<br />

of this syndrome with microdeletion of chromosome<br />

22q11 in 25% of cases. They also suggest that bronchomalacia<br />

is commonly associated with cardiomegaly and dilatation of<br />

the pulmonary artery and results in poor prognosis. 4 APVS, in<br />

the absence of VSD, is uncommon. As reported by Yeager<br />

et al, most of the cases presenting with an intact ventricular<br />

septum commonly reveal a patent ductus arteriosus, with<br />

relatively small pulmonary arteries and associated tricuspid<br />

atresia. 2 According to their study, the free communication<br />

Fig. 1 e An oblique coronal image of the fetal thorax reveals<br />

the aneurysmally dilated main pulmonary artery and<br />

absence of pulmonary valve echoes (arrow).<br />

Fig. 3 e Sagittal image of the fetal thorax reveals a large<br />

subaortic VSD (arrow) between the right ventricle (RV) and<br />

the left ventricle (LV). Aorta (AO) is seen arising from the LV.


indian heart journal 65 (<strong>2013</strong>) 191e193 193<br />

by Callan et al, the presence of polyhydramnios may indicate<br />

a poor prognosis. 5<br />

4. Conclusion<br />

We conclude that when a paracardiac cystic, pulsatile lesion is<br />

seen in the fetus in utero, APVS is an important differential<br />

diagnosis and other features associated with the syndrome,<br />

such as TOF and the presence or absence of the ductus arteriosus<br />

should be looked for.<br />

references<br />

Fig. 4 e CW Doppler interrogation of RVOT/MPA showing<br />

pulmonary regurgitation.<br />

between the ventricles and the aorta causes the blood flow to<br />

the atria to be reduced, while there is an increase in the<br />

ventricular end diastolic pressure; this may in turn affect the<br />

cardiac function and the development of the atrioventricular<br />

valve. Yeager et al also suggest that in the presence of a VSD<br />

these changes affect both the ventricles, thereby resulting in<br />

a poor prognosis, 2 as seen in our case. A grossly dilated<br />

pulmonary artery can cause compression of the tracheobronchial<br />

tree and the esophagus. This obstructs the normal<br />

amniotic fluid circulation, causing polyhydramnios. As stated<br />

1. Yaman C, Arzt W, Tulzer G, Tews G. Tetralogy of Fallot with<br />

absent pulmonary valve e prenatal diagnosis and<br />

management in the 2nd trimester. Geburtshilfe Frauenheilkd.<br />

1996;56:563e565.<br />

2. Yeager SB, Van Der Velde ME, Waters BL, Sanders SP. Prenatal<br />

role of the ductus arteriosus in absent pulmonary valve<br />

syndrome. Echocardiography. 2002;19:489e493.<br />

3. Zucker N, Rozin I, Levitas A, Zalzstein E. Clinical presentation,<br />

natural history and outcome of patients with absent<br />

pulmonary valve syndrome. Cardiol Young. 2004;14:402e408.<br />

4. Volpe P, Paladini D, <strong>Mar</strong>asini M, et al. Characteristics,<br />

associations and outcome of absent pulmonary valve<br />

syndrome in the fetus. Ultrasound Obstet Gynecol.<br />

2004;24:623e628.<br />

5. Callan NA, Kan JS. Prenatal diagnosis of tetralogy of Fallot<br />

with absent pulmonary valve. Am J Perinatol. 1991;8:15e17.


220<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

resolution (STR) and 30-day MACE (w7% in all groups)/stent<br />

thrombosis/bleeding were not significantly different between<br />

the randomized groups.<br />

Two of the strongest baseline determinants of infarct size<br />

are: 1) anterior MI location and 2) abnormal TIMI flow. This<br />

trial was limited to patients with proximal or mid LAD<br />

occlusion and TIMI 0e2 flow. Moreover, it only enrolled<br />

patients who could be treated early, in whom time window for<br />

effective myocardial salvage had not closed. The median time<br />

from symptom onset to hospital arrival was only 99 min and<br />

the median D-to-B time was 45 min. Thus the study population<br />

represents a highly selected cohort of patients with<br />

large anterior MI, in whom infarct size reduction should be<br />

feasible given early presentation and rapid treatment. Infarct<br />

size was assessed by cMRI, which strongly correlates with<br />

subsequent mortality. Unlike prior studies, which measured<br />

infarct size at 2e7 days (a period during which substantial<br />

myocardial edema is present, thereby interfering with<br />

assessment of viable myocardium) in this study cMRI was<br />

done at 30 days when much of myocardial edema had<br />

resolved.<br />

These results need to be placed in the context of previous<br />

studies. A meta-analysis of 6 RCT’s (1246 patients) reported<br />

enhanced survival with bolus intracoronary abciximab.<br />

However, the recent AIDA-STEMI trial (2065 patients) found<br />

nearly identical rates of MACE with bolus intracoronary and<br />

intravenous abciximab. However, this trial differs from these<br />

earlier studies in many ways: 1) unlike prior studies which<br />

included routine post-PCI intravenous abciximab infusion in<br />

both the groups, in this trial only bolus intracoronary abciximab<br />

was given in the randomized groups. 2) In all prior trials<br />

(including AIDA-STEMI), intracoronary abciximab was infused<br />

proximally through the guide catheter thereby limiting its<br />

penetration into occlusive thrombus and allowing spillage of<br />

the drug to LCx or backflow into the aorta. In contrast, the<br />

local drug delivery catheter (clearway catheter) used in this<br />

study achieves high intra-clot concentration of abciximab at<br />

the site of LAD occlusion and prolongs drug residence time,<br />

which may enhance platelet disaggregation and thrombus<br />

resolution. In the present study, an abciximab bolus delivered<br />

directly to the infarct lesion site (without a 12-hour infusion)<br />

reduced infarct size at 30 days in patients with anterior STEMI<br />

reperfused early.<br />

Regarding aspiration thrombectomy, in TAPAS, 1071<br />

patients with anterior and non-anterior STEMI who presented<br />

within 12 h of symptoms at a single-center were randomized<br />

to manual aspiration vs. no aspiration before primary PCI;<br />

aspiration resulted in modest improvements in MBG and STR<br />

but a marked reduction in 1-year mortality. Other trials have<br />

reported conflicting results, and in contrast to single-center<br />

studies, multicenter aspiration trials have been largely negative.<br />

Moreover, in TAPAS, aspiration did not reduce infarct<br />

size as measured by cardiac biomarkers, calling into question<br />

the mechanism underlying the survival benefit. The present<br />

multicenter trial, in which only patients presenting early with<br />

anterior MI and coronary anatomy optimal for aspiration were<br />

enrolled, and in which cMRI was used to assess infarct size at<br />

30 days was specifically designed to overcome many of the<br />

limitations from these earlier studies. The fact that manual<br />

thrombus aspiration did not reduce infarct size in this study<br />

makes a substantial clinical benefit unlikely, questioning its<br />

routine use in STEMI.<br />

9. Our opinion<br />

Regarding use of GPIIb/IIIa inhibitors: a) I/V bolus and infusion<br />

is to be discouraged because it achieves very little intra-clot<br />

concentration and also increases the risk of systemic bleeding.<br />

b) Only bolus intracoronary drug should be used, that too<br />

not into the guide catheter, but via a clearway catheter (we can<br />

use a simple PTCA balloon by making multiple holes on its<br />

surface, in case clearway catheter is not available).<br />

Regarding manual aspiration via Export catheter: a) the<br />

symptom onset to hospital arrival and the D-to-B time were<br />

substantially shorter in this study which is next to impossible<br />

in our context. b) As time passes by after STEMI thrombus<br />

tends to get organized and hence thrombus aspiration might<br />

have some role to play in late presenters of STEMI. However,<br />

the last word in this matter is yet to be written.<br />

Suraj Khanal*<br />

Assistant Professor of Cardiology, Department of Cardiology,<br />

3rd Floor, Block-C, Advanced Cardiac Center, PGIMER,<br />

Chandigarh 160012, India<br />

Ajay Bahl<br />

Associate Professor of Cardiology, PGIMER, Chandigarh, India<br />

*Corresponding author. Tel.: þ91 09878222526.<br />

E-mail address: khanal.s@rediffmail.com<br />

Azeem Latib, Antonio Colombo, Fausto Castriota,<br />

Antonio Micari, Alberto Cremonesi, Francesco De Felice,<br />

Alfredo <strong>Mar</strong>chese, Maurizio Tespili, Patrizia Presbitero,<br />

Gregory A. Sgueglia, Francesca Buffoli, Corrado Tamburino,<br />

Ferdinando Varbella, Alberto Menozzi, A randomized multicentre<br />

study comparing a paclitaxel drug-eluting balloon with<br />

a paclitaxel-eluting stent in small coronary vessels: The<br />

BELLO (Balloon Elution and Late Loss Optimization) study. J<br />

Am Coll Cardiol. 60 (2012) 2473e2480<br />

Objectives: The aim of this study was to evaluate the efficacy<br />

of drug-eluting balloons (DEB) compared with paclitaxel<br />

eluting stents (PES) for the reduction of restenosis in small<br />

vessels.<br />

Background: DEB have been shown to be effective in the<br />

treatment of coronary in-stent restenosis, but data are limited<br />

regarding their efficacy in de-novo disease.<br />

Methods: BELLO (Balloon Elution and Late Loss Optimization)<br />

is a prospective, multicentre trial that randomized 182 patients<br />

with lesions located in small vessels (reference diameter<br />


indian heart journal 65 (<strong>2013</strong>) 219e228 221<br />

(2.15 0.27 mm vs. 2.25 0.24 mm; p ¼ 0.003). The majority<br />

(89%) of lesions involved vessels with a diameter


indian heart journal 65 (<strong>2013</strong>) 142e146<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Coronary sinus filling time: A novel method to assess<br />

microcirculatory function in patients with angina and normal<br />

coronaries<br />

Vellani Haridasan a , Deepak Nandan a , Deepak Raju a , Gopalan Nair Rajesh a , C.G. Sajeev a ,<br />

Desabandhu Vinayakumar a , Kader Muneer a , Kadangot Babu a , M.N. Krishnan b, *<br />

a Govt. Medical College, Kozhikode, India<br />

b Professor and HOD, Department of Cardiology, Govt. Medical College, Kozhikode 673017, Kerala, India<br />

article info<br />

Article history:<br />

Received 21 October 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 21 February <strong>2013</strong><br />

Keywords:<br />

Syndrome X<br />

Coronary sinus filling time<br />

Coronary microcirculation<br />

abstract<br />

Objective: Dysfunction of the coronary microcirculation is considered as one of the factors<br />

responsible for symptoms and abnormal stress tests in patients with angina and normal<br />

coronaries (syndrome X). We sought to evaluate the usefulness of coronary sinus filling<br />

time (CSFT) to assess coronary microcirculation in this group of patients.<br />

Methods: We compared the CSFT of patients having definite angina or atypical angina with<br />

positive treadmill electrocardiography test (angina group), with that of patients undergoing<br />

coronary angiogram (CAG) prior to balloon mitral valvuloplasty (control group). During<br />

CAG, coronary sinus was visualized in appropriate views and CSFT in seconds was derived<br />

from frame count. Thrombolysis In Myocardial Infarction (TIMI) flow grade, corrected TIMI<br />

(cTIMI) frame count, TIMI Myocardial Perfusion grade (TMP) were assessed.<br />

Results: There were 41 patients in angina group and 16 in control group. Among the angina<br />

group 68.8% were females as against 81.8% in the control group. 87.8% (n ¼ 36) had typical<br />

angina. Mean CSFT was 4.25 0.72 s and 3.46 0.99 s in the angina group and control<br />

group respectively (p ¼ 0.001). No significant differences were found between the groups<br />

with respect to TMP (p ¼ 0.68) & cTIMI frame count (p ¼ 0.22).<br />

Conclusion: CSFT is a simple method to assess the transit time through coronary microcirculation.<br />

CSFT was significantly delayed in patients with angina and normal coronaries.<br />

TMP and cTIMI frame count were not significantly different between groups.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

1. Introduction<br />

Twenty percent of all diagnostic angiograms in patients<br />

referred for evaluation of chest pain have normal epicardial<br />

coronary arteries. 1 First described by Kemp in 1973, syndrome<br />

X comprises of a heterogenous group presenting with typical<br />

angina, a positive exercise stress test, normal epicardial coronaries<br />

and no clinical or angiographic evidence of epicardial<br />

coronary artery spasm. 2 Dysfunction of the coronary microcirculation<br />

may be one of the factors responsible for persistent<br />

anginal symptoms and abnormal stress test. 3,4 Prognosis of<br />

these patients is not as benign as reported by preliminary<br />

* Corresponding author. Tel.: þ91 4952356120, þ91 9846697553.<br />

E-mail addresses: kedaram@gmail.com, dr.mn.krishnan@gmail.com (M.N. Krishnan).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.002


indian heart journal 65 (<strong>2013</strong>) 142e146 143<br />

cohort studies. There are studies showing increased risk of<br />

myocardial infarction and cardiac death, especially in patients<br />

with positive stress test. 5,6 Women Ischemic Symptom Evaluation<br />

(WISE) trial showed that persistence of symptoms is<br />

associated with more than two-fold increase in cardiovascular<br />

events and concluded that such patients should undergo<br />

formal studies of vascular function and aggressive risk factor<br />

modification. 7 Noninvasive as well as invasive modes for<br />

assessing microcirculation have yielded inconsistent results<br />

and there is yet no simple method available at present to<br />

assess coronary microcirculation. 8e10 In this study, we have<br />

attempted to evaluate the usefulness of coronary sinus filling<br />

time (CSFT) for assessment of microcirculatory transit time in<br />

the coronary circulation.<br />

2. Patients and methods<br />

Adult patients presenting to the cardiology department of a<br />

major teaching hospital with angina-like chest pain from June<br />

2011 to January 2012 were screened for this study.<br />

The assessment of chest pain was done by two cardiologists<br />

of the department separately and doubtful cases were<br />

reviewed by a third cardiologist. Patients with chest pain were<br />

grouped into: 1) definite angina with/without positive TMT 2)<br />

probable angina with positive TMT and 3) non-cardiac chest<br />

pain. Definite (typical) angina was defined as substernal chest<br />

discomfort 1) of characteristic quality and duration 2) provoked<br />

by exertion or emotional stress 3) relieved by rest or<br />

sublingual nitrites. Probable angina was defined as those that<br />

met any two of the above characteristics; non-cardiac chest<br />

pain as those meeting one or none of the characteristics. 11<br />

Baseline evaluation, electrocardiogram (ECG) and echocardiography<br />

were done in all subjects. Definite and probable<br />

angina group underwent TMT. Patients were grouped as<br />

having either positive, negative or inconclusive test result<br />

based on standard criteria. 12 Patients with definite angina<br />

regardless of the result of TMT and patients with probable<br />

angina and a positive stress test were subjected to CAG.<br />

Among patients undergoing CAG, those with normal coronaries<br />

formed the angina group. The control group consisted<br />

of patients with mitral stenosis in sinus rhythm, normal coronary<br />

angiogram and normal left ventricular structure and<br />

function undergoing percutaneous balloon mitral valvuloplasty<br />

(BMV). The right atrial pressures and left ventricular<br />

end diastolic pressures were measured in all cases. Exclusion<br />

criteria were: 1) abnormal coronaries on angiography (any<br />

vascular irregularity/ectasia/stenoses), 2) patients with current<br />

or prior cardiovascular events by history, ECG or echocardiography,<br />

3) presence of cardiac diseases other than<br />

isolated mitral stenosis. Fig. 1 outlines the selection of patients<br />

for the study.<br />

Coronary angiogram was done with Philips Allura Xper<br />

FD20 (Philips Electronics, Eindhoven, The Netherlands) at a<br />

rate of 15 frames/s. Left coronary artery injection was taken<br />

with 7 mL contrast at 2 mL/s rate. A normal coronary artery<br />

was defined as epicardial coronary artery at angiography<br />

without any wall irregularities, ectasia or stenosis. TIMI<br />

Fig. 1 e Flow diagram showing the enrollment of patients for the study.


144<br />

indian heart journal 65 (<strong>2013</strong>) 142e146<br />

frame count, TMP grade and CSFT were obtained offline.<br />

CSFT was defined as the time taken in seconds for the<br />

contrast agent in the epicardial coronary artery to traverse<br />

the coronary microvasculature and reach the coronary sinus<br />

origin. CSFT was estimated as the difference between the<br />

frame count of maximum left anterior descending artery<br />

(LAD) system opacification at first diagonal (D 1 )/first septal<br />

(S 1 ) to that of the starting point of opacification of coronary<br />

sinus origin.<br />

Frame count at the maximum opacification of LAD at D 1 or<br />

S 1 , whichever was earlier (first frame count), was noted. A<br />

column of fully concentrated dye must extend across the<br />

entire width of LAD at D 1 /S 1 and move forwards. The frame<br />

count in which dye is first seen at the origin of coronary sinus<br />

is counted as the last frame and the frame count was noted<br />

(last frame count). Coronary sinus origin is defined as the<br />

point where great cardiac vein joins the posterolateral vein<br />

(that is marked by the joining of oblique vein of <strong>Mar</strong>shall to<br />

coronary sinus in whom it is seen). Coronary sinus was evaluated<br />

in at least two views, left anterior oblique (LAO) with<br />

appropriate cranial angulation and right anterior oblique<br />

(RAO) with caudal tilt. In these views, the tract and effluent<br />

were well visualized draining into the right atrium after<br />

sixeeight cycles on an average. The CSFT is calculated as:<br />

CSFT in seconds ¼ðlast frame count<br />

first frame count=15Þ<br />

TIMI Frame Count is defined as the number of angiographic<br />

frames elapsed from the first frame in which the contrast fully<br />

enters the artery to that in which it reaches the distal standardized<br />

landmark of the vessel of interest.<br />

For LAD, TIMI frame count [ Frame count at distal<br />

bifurcation e frame count at D 1 .<br />

The frame rates are corrected for the longer length of the<br />

LAD by dividing it by 1.7. 13 TMP is a simple descriptive score of<br />

myocardial opacification with contrast, distinct from the<br />

epicardial vessel, and provides a score of between 0 (no<br />

myocardial blush) and 3 (normal blush and clearance of dye<br />

from myocardium). 14,15 cTIMI frame count, TMP and CSFT in<br />

both the groups was compared and correlation analyzed.<br />

Analysis was done using two tailed t-test for equality of<br />

means. The demographic data was analyzed using Chi square<br />

test and Pearson correlation for correlation between CSFT<br />

frame count and cTIMI frame count.<br />

Table 1 e Baseline characteristics of patients and<br />

controls.<br />

Parameter<br />

21.84 5.0 in the control group p ¼ 0.224) or TMP (2.9 0.3 in<br />

angina group and 2.94 0.25 in control group p ¼ 0.68).<br />

4. Discussion<br />

Angina<br />

group n (%)<br />

Control<br />

group n (%)<br />

p value<br />

Sex<br />

M 13 (31.7) 3 (18.8)<br />

F 28 (68.3) 13 (81.2) 0.108<br />

Age e Mean (SD) 50.4 (7.6) 47.4 (9.6) 0.064<br />

Diabetes mellitus 16 (39) 2 (12.5) 0.053<br />

Hypertension 22 (53.7) 1 (6.2) 0.001<br />

Dyslipidemia 18 (43.9) 1 (6.2) 0.007<br />

Smoking 11 (26.8) 2 (12.5) 0.2<br />

Family history of CAD 6 (14.3) 0 0.11<br />

<strong>Heart</strong> rate e Mean (SD) 74 (12) 77 (14) 0.08<br />

RA pressure e Mean (SD) 3.4 (2.5) 5.3 (3.48) 0.06<br />

LV EDP 7 (3.6) 6 (2.4) 0.18<br />

Drugs<br />

1. Beta blockers 11 16 0.12<br />

2. CCB 8 2 0.04<br />

3. Statins 18 0 0.02<br />

4. Nitrates 32 0 0.003<br />

5. ACEI/ARB 18 4 0.03<br />

6. Aspirin 32 4 0.004<br />

RA ¼ Right atrium; LVEDP ¼ Left ventricular end diastolic pressure;<br />

CCB ¼ Calcium channel blocker; ACEI ¼ Angiotensin converting<br />

enzyme inhibitor; ARB ¼ Angiotensin receptor blocker.<br />

Our study demonstrated a delay in transit through microcirculation<br />

in patients with angina and normal coronaries. In a<br />

similar study, Sankareddi et al 16 have proposed that the time<br />

delay between LAD opacification and filling of coronary sinus<br />

would indicate the microcirculatory time. They went on to<br />

define CSFT as the difference in frame counts between<br />

maximum LAD opacification to the maximal opacification of<br />

the coronary sinus. They also calculated coronary sinus<br />

emptying time and coronary sinus emptying velocity as parameters<br />

to assess microcirculatory transit. In our study, we<br />

3. Results<br />

There were 41 cases in the angina group and 16 in the control<br />

group. Baseline data of both groups are given in (Table 1).<br />

Angina group and control group were compared with<br />

respect to following parameters e cTIMI frame, TMP Grade<br />

and CSFT. Mean CSFT frame count in angina group and in<br />

control group were 63.76 10.7 and 52.06 5.0 and mean CSFT<br />

were 4.2476 0.72 s and 3.4581 0.99 s, respectively (p ¼ 0.001)<br />

(Fig. 2). We could demonstrate a positive correlation between<br />

CSFT and cTIMI frame count in the angina group (correlation<br />

coefficient ¼ 0.7) (Fig. 3).<br />

No significant differences were found between the two<br />

groups with regard to cTIMI (23.97 6.2 in angina group and<br />

Fig. 2 e Coronary sinus filling time in angina group and<br />

control group.


indian heart journal 65 (<strong>2013</strong>) 142e146 145<br />

Fig. 3 e Correlation of cTIMI frame count and CSFT.<br />

included only CSFT, since coronary sinus emptying time and<br />

velocity depend on the drainage characteristics of coronary<br />

sinus rather than that of the coronary microcirculation. We<br />

modified the definition of CSFT as the difference in frame<br />

counts between maximum LAD opacification to the beginning<br />

of filling of coronary sinus origin. The coronary sinus is a<br />

venous conduit, which drains between 80% and 85% of unsaturated<br />

blood of the left ventricle. 17 Studies have shown<br />

that there is dynamic variation of the coronary sinus lumen<br />

during normal cardiac cycle and has been categorized into<br />

passive, normal response and hyperactive. 18 An exaggerated<br />

response to this mechanism could potentially modify cyclic<br />

coronary circulation and perfusion, causing slow flow phenomena.<br />

19 Modulation of coronary sinus outflow pressure in<br />

normal canine hearts affects intramyocardial tissue pressure<br />

and has been shown to reduce blood flow in the subepicardial<br />

tissue layer independently of coronary artery pressure. 20<br />

Since the maximal opacification of the coronary sinus would<br />

depend on multiple factors other than the condition of the<br />

microcirculation, we decided to assess the filling at the origin<br />

of coronary sinus. Our study agrees with the previous study<br />

that CSFT is delayed in angina with normal coronaries. Our<br />

study had a larger study population (n ¼ 41) compared to the<br />

previous study (n ¼ 10). We also compared other parameters of<br />

perfusion, cTIMI frame count and TMP. These two parameters<br />

were not significantly different between angina group and<br />

control group as in other studies. 14,21,22 In our study there was<br />

a positive correlation between cTIMI frame count & CSFT in<br />

the case group (r ¼ 0.730). There was a trend toward delayed<br />

CSFT in those patients with an increased cTIMI frame count.<br />

Mahfouz et al could demonstrate a marked delay in cTIMI<br />

frame count in a group of syndrome X patients, even though<br />

other studies have yielded mixed results. 23 A few studies<br />

could demonstrate abnormal TMP as a surrogate marker for<br />

dysfunctional coronary microvasculature in syndrome X. 24<br />

We could not demonstrate any correlation between TMP and<br />

CSFT in this study.<br />

We have not been able to find any study examining the<br />

relation between CSFT and long-term outcomes; nor could we<br />

find any study on the effect of coronary dilators like adenosine<br />

or dipyridamole on CSFT. It would be worthwhile to study the<br />

differential power of CSFT on long-term outcomes in patients<br />

with angina and normal coronaries.<br />

There were several limitations in our study, as this was one<br />

of the pilot studies of CSFT. One of the limitations of this study<br />

was the small number of control population. We could not<br />

recruit absolutely normal subjects as control population<br />

because of ethical issues. We took mitral stenosis patients<br />

undergoing CAG prior to BMV as controls. In case of control<br />

population, we had to exclude patients with mean pulmonary<br />

artery pressure 25 mmHg, as elevated right atrial pressures<br />

prolonged coronary sinus filing time. Another limitation was<br />

that coronary spasm was not excluded. We did not standardize<br />

the injection speed and force as we used hand injection;<br />

this could have affected the results. However, Gibson<br />

et al have shown that the difference in hardware, injection<br />

methods and phase of cardiac cycle did not produce much<br />

variability in corrected frame count calculation. 13 Since we did<br />

not come across any study that used automated injectors for<br />

standardization our study used manual injections at prespecified<br />

rate and volume. Our study group and controls have<br />

a preponderance of female subjects; thus it is not clear<br />

whether the conclusions can be extended to male patients.<br />

CSFT may be proposed as a method to risk-stratify patients<br />

with angina and normal coronaries. Moreover, CSFT may be<br />

used as a simple and quantitative test in percutaneous coronary<br />

intervention setting to assess myocardial reperfusion.<br />

Further studies are required to evaluate this proposition.<br />

5. Conclusion<br />

CSFT is a simple method to assess the transit time through<br />

coronary microcirculation. CSFT was significantly delayed in<br />

patients with angina and normal coronaries. TMP and cTIMI<br />

frame count did not differ significantly between the groups<br />

studied.<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Chierchia SL, Fragasso G. Angina with normal coronary<br />

arteries: diagnosis, pathophysiology and treatment. Eur <strong>Heart</strong><br />

J. 1996;17:14e19.<br />

2. Kemp HG. Left ventricular function in patients with angina<br />

syndrome and normal coronary arteriograms. Am J Cardiol.<br />

1973;32:375e376.<br />

3. Opherk D, Zebe H, Weihe E, et al. Reduced coronary dilatory<br />

capacity and ultrastructural changes of the myocardium in<br />

patients with angina pectoris but normal coronary<br />

arteriograms. Circulation. 1981;63:817e825.


146<br />

indian heart journal 65 (<strong>2013</strong>) 142e146<br />

4. Cannon III RO, Canici PG, Epstein SE. Pathophysiological<br />

dilemma of syndrome X. Circulation. 1992;85:883e892.<br />

5. Prognosis in women with myocardial ischemia in the absence<br />

of obstructive CAD: results from the National Institute of<br />

Health-National <strong>Heart</strong>, Lung, & Blood Institute e Sponsored<br />

Women Ischemic Syndrome Evaluation Trial (WISE).<br />

Circulation. 2004;109:2993.<br />

6. Bugiardini R. Women, “non specific” chest pain, normal or<br />

near normal CAG are not synonymous with favourable<br />

outcome. Eur <strong>Heart</strong> J. 2006;27:1387.<br />

7. Johnson BD, Pepera CJ, Reis SE, et al. Persistent chest pain<br />

predicts cardiovascular events in women without obstructive<br />

CAD: results from NIH-NHLBI-sponsored Women Ischemic<br />

Syndrome Evaluation (WISE) study. Eur <strong>Heart</strong> J. 2006;27:1408.<br />

8. Meller J, Goldsmith SJ, Rudin A, et al. Spectrum of exercise<br />

thallium-201 myocardial perfusion imaging in patients with<br />

chest pain and normal coronary angiograms. Am J Cardiol.<br />

1979;43:717e723.<br />

9. Rossetti E, Fragasso G, Vanzulli A, et al. Magnetic resonance<br />

contrast enhancement with gadolinium-DTPA in patients<br />

with angina and angiographically normal coronary arteries:<br />

effect of chronic beta-blockade. Cardiologia.<br />

1999;44(7):653e659.<br />

10. Dijkmans PA, Knaapen P, Sieswerda GT, et al. Quantification<br />

of myocardial perfusion using intravenous myocardial<br />

contrast echocardiography in healthy volunteers: comparison<br />

with positron emission tomography. J Am Soc<br />

Echocardiography. 2006;19:285e293.<br />

11. ACC/AHA updated guidelines for management of patients<br />

with stable chest pain syndromes and known or suspected<br />

ischemic heart disease. J Am Coll Cardiol. 2003;41(1):159e168.<br />

12. A report of the ACC/AHA Taskforce on practice guidelines<br />

(Committee to update the 1997 exercise testing guidelines).<br />

Circulation. 2002;106:1883e1892.<br />

13. Gibson CM, Cannon CP, Daley WL, et al. TIMI frame count: a<br />

quantitative method of assessing coronary artery flow.<br />

Circulation. 1996;93:879e888.<br />

14. Bertomeu-Gonzáleza Vicente, Bodía Vicent, Sanchis Juan.<br />

Limitations of myocardial blush grade in the evaluation of<br />

myocardial perfusion in patients with acute myocardial<br />

infarction and TIMI grade 3 flow. Esp Cardiol. 2006;59:<br />

575e581.<br />

15. Dibra A, Mehilli J, Dirschinger J, et al. Thrombolysis in<br />

myocardial infarction myocardial perfusion grade in<br />

angiography correlates with myocardial salvage in patients<br />

with acute myocardial infarction treated with stenting or<br />

thrombolysis. J Am Coll Cardiol. 2003;41:925e929.<br />

16. Sangareddi V, Alagesan R. Coronary Sinus Filling and<br />

Emptying Time: A New Parameter to Assess Coronary<br />

Microcirculation by a Simple Angiographic Frame Count; 59th<br />

Annual Conference of the Cardiological Society of India<br />

December 7e10, 2008 [abstract].<br />

17. Singh Jagmeet P, Houser Stuart, Heist E Kevin, Ruskin Jeremy<br />

N. The coronary venous anatomy: a segmental approach to<br />

aid cardiac resynchronization therapy. J Am Coll Cardiol.<br />

2005;46:68e74.<br />

18. Barcelo A, De La Fuente LM, Stertzer SH. Anatomic and<br />

histologic review of the coronary sinus. Int J Morphol.<br />

2004;22(4):331e338.<br />

19. Krakover R, Blatt A, Hendler A, et al. Angiographic functional<br />

characterization of the coronary sinus. Isr Med Assoc J.<br />

2005;7:374e376.<br />

20. Rouleau JR, White M. Effects of coronary sinus pressure<br />

elevation on coronary blood flow distribution in dogs with<br />

normal preload. Can J Physiol Pharmacol. 1985;63:787e797.<br />

21. Chen YC, Hou CJ, Tsai CH, et al. Relationships of the<br />

thrombolysis in myocardial infarction frame count with<br />

clinical, hemodynamic and medicine variables in syndrome X<br />

patients. Int J Gerontol. 2008;2(3):109e114.<br />

22. Nihat S, Yusuf T, Ugurii H, et al. Coronary blood flow in<br />

patients with cardiac syndrome X. Coron Artery Dis.<br />

2007;18(1):45e48.<br />

23. Mahfouz Ragab A, Abdou Mohamed, Elsaeed Ashraf,<br />

Kandil Nader T. Cardiac syndrome-X: is it benign or malignant?<br />

An Egyptian follow-up study. Open J Cardiol. 2011;2:1.<br />

24. Atmaca Y, Zdemir AO. Angiographic evaluation of myocardial<br />

perfusion in patients with syndrome X. Am J Cardiol.<br />

2005;96(6):803e805.


indian heart journal 65 (<strong>2013</strong>) 229e231<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Arrhythmia Graphics<br />

Nodal and infranodal atrioventricular conduction block:<br />

Electrophysiological basis to correlate the ECG findings<br />

Bharat K. Kantharia a,b,c, *, Arti N. Shah d<br />

a Professor of Medicine, The University of Texas-Health Science Center at Houston, 6431 Fannin Street, Suite MSB 1.246, Houston,<br />

TX 77030, USA<br />

b Director, Clinical Cardiac Electrophysiology Fellowship Training Program, 6431 Fannin Street, Suite MSB 1.246, Houston, TX 77030, USA<br />

c Director, Cardiac Electrophysiology Laboratories-Memorial Hermann Hospital, 6431 Fannin Street, Suite MSB 1.246, Houston,<br />

TX 77030, USA<br />

d Elmhurst Hospital Center, Mount Sinai School of Medicine, NY, USA<br />

article info<br />

Article history:<br />

Received 2 January <strong>2013</strong><br />

Accepted 14 February <strong>2013</strong><br />

Available online 24 February <strong>2013</strong><br />

Keywords:<br />

HisePurkinje system<br />

Atrioventricular node<br />

2:1 block<br />

Atrioventricular nodal block<br />

Infra-Hisian block<br />

abstract<br />

A 68-year-old woman with a history of dilated non-ischemic cardiomyopathy presented<br />

with syncope. The index ECG showed sinus rhythm with left bundle branch block.<br />

On telemetry episodes of sinus rhythm with narrower QRS complexes conduced in 2:1<br />

pattern were noted. Invasive electrophysiological study was performed to determine cause<br />

of syncope. Normal conduction up to the AV node with an AH interval of 79 ms<br />

(normal ¼ 55e125 ms) was observed. However, every alternate sinus beat was blocked after<br />

the inscription of His deflection (infra-Hisian block). The narrow beats conducted through<br />

the His bundle with HV intervals of 54 ms (normal ¼ 35e55 ms). When 1:1 conduction<br />

resumed further abnormality of the HisePurkinje conduction system became evident with<br />

a QRS morphology that of an LBBB and prolongation of HV interval (HV ¼ 96 ms). Criteria to<br />

differentiate nodal versus infranodal block based on electrophysiological properties of the<br />

nodal and infranodal system are discussed.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

A 68-year-old woman with a history of dilated non-ischemic<br />

cardiomyopathy (left ventricular ejection fraction 40%), presented<br />

with syncope. Her physical examination was unremarkable.<br />

The index ECG showed sinus rhythm with left<br />

bundle branch block (LBBB). On telemetry episodes of sinus<br />

rhythm with narrower QRS complexes conduced in 2:1 pattern<br />

were noted. Invasive electrophysiological (EP) study was performed<br />

to determine cause of syncope.<br />

Relevant observations made during EP study are as shown<br />

in Fig. 1. Panels A and B: 12 lead ECGs are recorded at a paper<br />

speed of 25 mm/s. The baseline rhythm is sinus at a rate of 88<br />

beats per minute (bpm) [sinus cycle length of 680 ms (ms)] that<br />

has 1 to 1 atrioventricular (AV) conduction with QRS<br />

morphology of LBBB, PR intervals of 200 ms and QRS durations<br />

of 120 ms (Panel A). The rhythm spontaneously converts to<br />

sinus with 2:1 AV block (Panel B). During the 2:1 AV block<br />

rhythm, the conducted beats have PR intervals of 200 ms and<br />

the QRS complexes are narrower and measure 100 ms in<br />

duration. The intervals between the two consecutive P waves<br />

(PeP interval) remain fixed at 680 ms, The intervals between<br />

* Corresponding author. Tel.: þ1 713 500 6590; fax: þ1 713 500 6556.<br />

E-mail addresses: bharat.k.kantharia@uth.tmc.edu, bkantharia@yahoo.com (B.K. Kantharia).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.003


230<br />

indian heart journal 65 (<strong>2013</strong>) 229e231<br />

Fig. 1 e Surface 12-lead ECGs (A and B) and the tracings of intracardiac electrograms (C and D) during AV block.<br />

the R wave to the next conducted P wave (R to P þ 1 interval)<br />

remain fixed at 1160 ms. Panels C and D: The tracings are<br />

recorded at a paper speed of 50 mm/s and 100 mm/s, respectively.<br />

From top to bottom, the tracings show surface ECG<br />

leads I, aVF and V1, intracardiac electrograms from the high<br />

right atrium (hRA) corresponding to the P waves, the AV nodal<br />

junction region covering the proximal, mid and distal His<br />

bundle (HISp, HISm and HISd, respectively) corresponding to<br />

the PR intervals, and the outflow tract region of the right<br />

ventricle (RVot) corresponding to the R waves. Each sinus beat<br />

is conducted normally up to the AV node with an AH interval<br />

of 79 ms (normal ¼ 55e125 ms). However, every alternate<br />

sinus beat is blocked after the inscription of His deflection<br />

(infra-Hisian block). The narrow beats conduct through the<br />

His bundle with HV intervals of 54 ms (normal ¼ 35e55 ms).<br />

When 1:1 conduction resumes (the last beat in Panel D) further<br />

abnormality of the HisePurkinje conduction system becomes<br />

evident with a QRS morphology that of an LBBB and prolongation<br />

of HV interval (HV ¼ 96 ms).<br />

1. Commentary<br />

On routine ECGs, right bundle branch block (RBBB) may be<br />

seen even in otherwise perfectly healthy individuals. On the<br />

other hand, the inscription of LBBB usually indicates abnormality<br />

of the HisePurkinje system, with the exception of LBBB<br />

that may be observed during rapid supraventricular tachycardia<br />

with aberrant conduction secondary to rate related<br />

functional conduction block/delay in the bundle.<br />

By ECG criteria alone, it can be very hard to determine the<br />

level of block, i.e. Mobitz I (AV nodal) or Mobitz II (infranodal)<br />

block in the presence of 2:1 AV conduction block. Although, for<br />

the most part wide QRS complexes suggest infranodal block,<br />

and narrow QRS complexes indicate AV nodal block, these<br />

criteria are not reliable. In the presence of 2:1 AV conduction<br />

block, if the PR intervals of the conducted beats remain short<br />

and fixed, the level of block may be considered infranodal<br />

(Mobitz II block). On the other hand, if the PR intervals of the<br />

conducted beats are longer and not fixed, the level of block<br />

may be considered AV nodal (Mobitz I block). In a typical<br />

Mobitz I (AV nodal Wenckebach) block, the PR interval of the<br />

first conducted complex after the dropped beat shortens due<br />

to recovery of normal AV nodal conduction. If the AV<br />

Wenckebach periodicity were to continue then the next PR<br />

interval would prolong considerably. The subsequent complexes<br />

although would have further prolongation of the PR<br />

intervals, the increments compared to the preceding intervals<br />

are less. Thus, the RR intervals may demonstrate apparent<br />

shortening with prolongation of the PR intervals. In case of 2:1<br />

AV block, if the PR intervals of the conducted beats remain<br />

short and fixed then it would be difficult to conceptualize that<br />

the AV node after conducting a sinus beat normally (normal<br />

PR interval) would impart so much conduction delay for the


indian heart journal 65 (<strong>2013</strong>) 229e231 231<br />

very next sinus beat at an identical sinus cycle length that the<br />

beat would block within the AV node. However, in case of the<br />

HisePurkinje system dysfunction, conduction of sinus beats<br />

through the infranodal structures may be totally unreliable<br />

and only slight decrement in the conduction through the<br />

HisePurkinje system may result in total conduction block.<br />

On a surface ECG, prolongation of the PR interval and<br />

correspondingly a longer AH interval during EP study may be<br />

observed if there is a rapid input to the AV node by a shortcoupled<br />

premature beat that conducts through the AV node<br />

with decremental conduction physiology. Likewise, with recovery<br />

of the AV nodal conduction, shorter PR and AH intervals<br />

may be observed if there is a delayed input to the AV<br />

node by a long-coupled premature beat. Thus, during 2:1 AV<br />

block, if there is a reciprocal relationship between the R to<br />

P þ 1 interval and subsequent PR interval (shorter R to P þ 1<br />

interval resulting in a longer subsequent PR interval and vice<br />

versa) is observed then the level of block is most likely within<br />

the AV node. On the other hand, if the conduction block<br />

occurs in the His-Purkinje system (infranodal block) then<br />

irrespective of the variations in the R to P þ 1 interval, the<br />

subsequent PR intervals remain fixed.<br />

Of course, certain maneuvers and interventions to alter the<br />

vagal and sympathetic balance may help to differentiate AV<br />

nodal block from block in the infranodal HisePurkinje system.<br />

For example, although the carotid sinus massage which increases<br />

the vagal tone and slows the sinus rate and conduction<br />

through the AV node would further deteriorate nodal<br />

block, it may improve the infranodal block by improving the<br />

His-Purkinje conduction of slower sinus beats. Exercise,<br />

atropine and isoproterenol would improve nodal block but<br />

would deteriorate infranodal block.<br />

Conflicts of interest<br />

All authors have none to declare.


indian heart journal 65 (<strong>2013</strong>) 198e200<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Case Report<br />

Constrictive pericarditis following Coronary Artery Bypass<br />

Grafting<br />

Anirban Kundu a, *, Om Prakash Yadava a , Arvind Prakash b<br />

a Department of Cardiac Surgery, National <strong>Heart</strong> Institute, New Delhi, India<br />

b Department of Cardiac Anesthesiology, National <strong>Heart</strong> Institute, New Delhi, India<br />

article info<br />

Article history:<br />

Received 9 July 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 21 February <strong>2013</strong><br />

Keywords:<br />

Constrictive pericarditis<br />

Coronary Artery Bypass Grafting<br />

Thoracotomy<br />

Pericardiectomy<br />

abstract<br />

Constrictive pericarditis following Coronary Artery Bypass Surgery is an uncommon disorder.<br />

We report a patient who developed constrictive pericarditis after Coronary Artery<br />

Bypass Grafting. After an unsuccessful trial of medical management and pericardial tapping,<br />

he underwent pericardiectomy via a left posterolateral thoracotomy.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

1. Case report<br />

The patient was a 66-year-old normotensive, non-diabetic<br />

gentleman with normal left ventricular function, who underwent<br />

off-Pump CABG in January 2009, wherein he<br />

received three grafts. However, during sternal closure he had<br />

a sudden and unexplained hemodynamic collapse, necessitating<br />

emergent cardiopulmonary bypass (CPB) support. His<br />

subsequent post-operative course was uneventful and he was<br />

discharged on the 7 th post-operative day. Pre-discharge echo<br />

showed minimal pericardial effusion posterolateral to the left<br />

ventricle (LV). After 16 months, he presented with shortness of<br />

breath, pedal edema and easy fatigability. 2D echocardiography<br />

revealed biatrial dilatation, small LV, marked pericardial<br />

effusion with fibrous strands, no constriction, marked thickening<br />

of posterolateral pericardium, normal LV ejection fraction<br />

and raised LVEDP. 300 ml of hemorrhagic fluid was<br />

drained under echocardiographic guidance. However, he<br />

again presented with similar complaints after six months.<br />

This time, echocardiography showed a large pericardial<br />

collection causing posterolateral compression of the LV, with<br />

pericardium thickened to 7 mm and features of constrictive<br />

physiology. MRI of thorax showed a mass 8 cm in diameter<br />

compressing the LV. Keeping in mind the clinical and imaging<br />

findings and the history of recurrence, it was decided to<br />

operate. He underwent preoperative cardiac catheterization<br />

and coronary angiography, which confirmed the echocardiographic<br />

findings and demonstrated patent grafts. Based on the<br />

MRI findings which suggested a left lateral compression of the<br />

LV (Fig. 2), and the presence of patent grafts, our surgical<br />

approach was left posterolateral thoracotomy. There was a<br />

large pleural effusion of about 2 L of straw colored fluid. The<br />

LV was encased by 1 cm thick pericardium with 500 ml<br />

of altered blood and partially-organized clots (Fig. 1). Pericardiectomy<br />

over the LV was done with thorough removal of<br />

the clots and fluid, leaving a strip of pericardium along the<br />

* Corresponding author. Tel.: þ91 11 46600700 (Ext: 4098), þ91 11 46600700 (Ext: 4055); fax: þ91 11 2642 8372.<br />

E-mail addresses: dockundu@yahoo.com, drkundu@indiatimes.com, onku2@rediffmail.com (A. Kundu).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.004


indian heart journal 65 (<strong>2013</strong>) 198e200 199<br />

Fig. 1 e Operative photograph showing partially opened<br />

thickened pericardium.<br />

phrenic nerve. Post-operative echocardiography showed mild<br />

posterolateral pericardial collection with no constriction and<br />

the patient was discharged after 4 days.<br />

2. Discussion<br />

Constrictive pericarditis as a complication following CABG is<br />

rare and was first reported in 1972. 1 Constrictive pericarditis is<br />

caused by adhesions and fibrous contracture of the pericardial<br />

sac. This impairs normal diastolic ventricular filling, causing a<br />

fall in stroke volume and elevation of systemic and pulmonary<br />

venous pressures. Fatigue, dyspnea and signs of congestive<br />

cardiac failure result. At our institution, at which almost 500<br />

major cardiac surgical proceduresdmainly CABGs e are performed<br />

annually; this was the first such case. While it is true<br />

that such cases eventually requiring pericardiectomy are rare,<br />

the true incidence of pericardial constriction post-surgery may<br />

be underestimated. Mild cases may mimic low cardiac output<br />

syndrome, with low systemic pressures and high venous<br />

pressures, especially post-valve surgeries, or following CABG<br />

with severe LV dysfunction. Patients with mild disease may<br />

respond well to diuretic treatment given for heart failure and<br />

thus never undergo investigations to establish the diagnosis,<br />

which requires a high degree of clinical suspicion. 2 Definitive<br />

factors that predispose to the development of constriction in<br />

patients after cardiac surgery are still not well known. Studies<br />

on animal models have demonstrated that pericardial adhesions<br />

will develop if spilled blood comes into contact with an<br />

injured serosal surface. 3 The volume of blood spillage during<br />

surgery is variable, but the initiation of inflammation and<br />

fibrosis depends on the presence of pericardial damage. Postpericardiotomy<br />

syndrome (PPS) has been postulated as a<br />

potential cause and has been found to occur in up to 30% of<br />

patients after cardiac surgery. 4 The presence of PPS does not<br />

necessarily predict which patients will develop constriction,<br />

and its absence does not preclude the development of<br />

constriction. However, PPS is an indication of inflammation in<br />

and around the pericardium, and perhaps these patients<br />

should be watched more closely for development of constrictive<br />

pericarditis. Definitive management of this condition remains<br />

re-do surgery with pericardiectomy. Patients who have<br />

undergone previous surgery, especially CABG, pose a greater<br />

risk with patent grafts lying just beneath the sternum, which<br />

are susceptible to injury during the procedure. Lee et al have<br />

recommended a non-midline sternotomy approach, to avoid<br />

damage to patent grafts. Patients in their series underwent<br />

pericardial stripping via right, left and bilateral thoracotomies,<br />

depending on the extent of constriction. 5 Our patient underwent<br />

CABG over two years ago, which had required CPB support.<br />

It is well known that CPB is associated with increased risk<br />

of bleeding complications and inflammatory response.<br />

Removal of drainage tubes following CABG might have been<br />

followed by gradual oozing from the raw surfaces caused by<br />

the surgery, which could have led to inflammation, fibrosis<br />

and constriction. Imaging studies revealed a marked thickening<br />

on the left lateral aspect of the heart. This correlates<br />

with the post-operative echocardiography findings of a<br />

posterolateral collection. Keeping in mind the site of<br />

compression and the fact that the patient was post-CABG with<br />

patent grafts, it was decided to use a left posterolateral thoracotomy.<br />

Apart from obviating the need for any extra precaution<br />

with regard to the grafts, this approach avoided the<br />

morbidity associated with a re-do sternotomy and very<br />

importantly, allowed for a satisfactory decompression of the<br />

LV, which was bearing the brunt of the constrictive process.<br />

3. Conclusion<br />

Fig. 2 e Preoperative MRI showing thickened pericardium.<br />

Based on the experience of this case, we conclude that pericardiectomy<br />

via lateral thoracotomy is a safe and effective<br />

approach in cases of post-CABG constrictive pericarditis, in<br />

view of the presence of patent grafts, and added morbidity of a<br />

re-do sternotomy. Also, this uncommon complication has to<br />

be considered in all post-cardiac surgery patients presenting<br />

with signs of unexplained myocardial failure, before ascribing<br />

them to the underlying cardiac disorder, or to post-cardiac<br />

surgery LV dysfunction.


200<br />

indian heart journal 65 (<strong>2013</strong>) 198e200<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Kendall ME, Rhodes GR, Wolfe W. Cardiac constriction<br />

following aorta to coronary bypass surgery. J Thorac Cardiovasc<br />

Surg. 1972;64:142e153.<br />

2. Killian DM, Furiasse JG, Scanlon PJ, et al. Constrictive<br />

pericarditis after cardiac surgery. Am <strong>Heart</strong> J.<br />

1989;118:563e568.<br />

3. Cliff WJ, Grobetz J, Ryan GB. Postoperative pericardial<br />

adhesions. The role of mild serosal injury and spilled blood. J<br />

Thorac Cardiovasc Surg. 1973;65:744e750.<br />

4. Drusin LM, Engle MA, Hagstrom JWC, et al. The<br />

postpericardiotomy syndrome. A six year epidemiologic study.<br />

N Engl J Med. 1965;272:597e602.<br />

5. Lee C, Yang S, Yang H, et al. Pericardiectomy through a right<br />

anterior thoracotomy for constrictive pericarditis after<br />

coronary artery bypass grafting. J Med Sci. 2007;27(2):085e088.


indian heart journal 65 (<strong>2013</strong>) 194e197<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Case Report<br />

Intravascular ultrasound-guided revascularization of a<br />

chronically occluded left main coronary artery<br />

Alberto Ranieri De Caterina, Florim Cuculi, Adrian P. Banning*<br />

Oxford <strong>Heart</strong> Centre, Oxford University Hospitals, Headley Way, Oxford OX3 9DU, UK<br />

article info<br />

Article history:<br />

Received 19 September 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 21 February <strong>2013</strong><br />

Keywords:<br />

Left main coronary artery<br />

Chronic total occlusion<br />

Intravascular ultrasound<br />

abstract<br />

We describe a case of a left main coronary artery (LMCA) chronic total occlusion (CTO),<br />

which we elected to treat through percutaneous coronary intervention (PCI). In this case<br />

report, we briefly review the prevalence of LMCA CTO, discuss the feasibility of PCI versus<br />

surgical revascularization and highlight the importance of intravascular ultrasound in the<br />

guidance of these complex procedures.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

1. Background<br />

Chronic total occlusion (CTO) of the left main coronary artery<br />

(LMCA) is a rare finding in everyday practice. Here we describe<br />

a successful percutaneous revascularization of a LMCA CTO in<br />

a patient presenting with mild-to-moderate symptoms. Specifically,<br />

in this case we highlight the importance of intravascular<br />

ultrasound in the guidance of these complex procedures.<br />

2. Case presentation<br />

A 75-year-old lady was referred to our heart centre for the<br />

evaluation of breathlessness to minimal exertion, which had<br />

suddenly worsened after an episode of “unwellness” three<br />

months before. She denied chest pain, was overweight (Body<br />

Mass Index, BMI, of 43 kg/m 2 ) and had a history of<br />

well-controlled hypertension and hypercholesterolemia. An<br />

echocardiogram showed mildly impaired global left ventricular<br />

(LV) contractility (LV ejection fraction 46%) with hypokinetic<br />

anterior wall. Single photon emission computed<br />

tomography (SPECT) revealed severely impaired perfusion in<br />

the left anterior descendent (LAD) territory and inducible<br />

ischemia in the lateral wall. At coronary angiography an ostial<br />

LMCA occlusion with some antegrade vessel opacification<br />

through a microchannel and retrograde filling from a dominant<br />

right coronary artery (RCA) was found (Fig. 1). Syntax<br />

score was 27, while Euroscore II was 2.55%.<br />

The patient underwent percutaneous coronary intervention<br />

(PCI) through bilateral femoral approach for contralateral<br />

injection if needed. LMCA ostium was engaged with a Judkins<br />

left 4 6F guide catheter and the lesion successfully crossed<br />

engaging the microchannel with a Pilot 50 wire (Abbott<br />

Vascular, Canada). After multiple predilations with 1.5 and 2.5<br />

balloons, an intravascular ultrasound (IVUS) run from the<br />

proximal circumflex artery (Cx) was performed showing<br />

* Corresponding author. Tel.: þ44 1865 228934; fax: þ44 1865 220585.<br />

E-mail address: Adrian.Banning@ouh.nhs.uk (A.P. Banning).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.005


indian heart journal 65 (<strong>2013</strong>) 194e197 195<br />

Fig. 1 e LMCA occlusion. Angiography shows eterocoronary retrograde vessel filling from RCA (panel A) and minimal<br />

antegrade filling (panel B, arrows).<br />

diffuse concentric atheroma extending from bifurcation to<br />

ostial LMCA (Fig. 2) with a native vessel diameter of 3.9 mm. A<br />

4 24 mm drug eluting stent (Promus Element, Boston<br />

Scientific) from proximal Cx to ostial LMCA was successfully<br />

implanted (Fig. 3, left panel). Post-stenting IVUS run from<br />

proximal LAD revealed mild ostial LAD disease, suboptimal<br />

stent struts expansion and coverage of LMCA ostium with<br />

minimal stent protrusion into aorta (Fig. 3, mid panels). Final<br />

kissing balloon with 4.0 12 mm (to Cx) and 3.0 12 mm<br />

(to LAD) non-compliant (NC) balloons (Fig. 3, right panel)<br />

followed by stent expansion of the body of LMCA with a<br />

5 8 mm NC balloon was performed. Excellent angiographic<br />

result was achieved and a final IVUS run confirmed optimal<br />

stent expansion (Fig. 4). The recovery after the procedure<br />

was uneventful, the patient was discharged the day after<br />

and was asymptomatic at clinical follow-up.<br />

3. Discussion<br />

Fig. 2 e LMCA recanalization. Angiographic (panel A) and<br />

IVUS (panel B and C) appearance after multiple balloon<br />

LMCA predilation. Diffuse circumferential atheroma is<br />

evident both at proximal (panel B) and mid left main<br />

coronary artery (panel C).<br />

CTO of the LMCA is a rare condition, with a reported prevalence<br />

ranging from 0.04 to 0.4%. 1,2 Two reasons can explain such a<br />

low prevalence. On the one hand, stable severe LMCA disease is<br />

almost always symptomatic, so that patients can be often<br />

diagnosed and treated before further progression. On the other<br />

hand, acute LMCA occlusion is frequently fatal and survival is<br />

possible only in patients with a dominant RCA providing sufficient<br />

collateral formation. Interestingly, our patient suffered<br />

mainly from shortness of breath and did not complain of<br />

angina, although SPECT demonstrated considerable inducible<br />

ischemia. Although this is not uncommon in diabetic patients<br />

it is surprising in a non-diabetic patient with LMCA occlusion.<br />

Although the recommendation for LMCA revascularization<br />

is still coronary artery bypass grafting (CABG), emerging evidence<br />

supports the feasibility of PCI in this setting. 3,4 Specifically,<br />

the pre-specified LMCA subgroup in the Syntax trial<br />

showed no significant difference for safety and efficacy endpoints<br />

compared to CABG at 1-year follow-up. 4 In the case here<br />

presented, although the high Syntax score would have<br />

indicated surgical revascularization, the poor functional<br />

status (NYHA III class) and severe obesity (BMI > 40 kg/m 2 )<br />

suggested PCI as first attempt. In addition, PCI was a low risk<br />

option compared to surgery and we were reassured by<br />

option of surgical revascularization in case of failure. We<br />

also felt that PCI could provide the potential advantage of the<br />

reestablishment of normal coronary anatomy.


196<br />

indian heart journal 65 (<strong>2013</strong>) 194e197<br />

Fig. 3 e IVUS-guided PCI optimization. After Cx-LMCA stenting (left panel) IVUS run shows suboptimal stent expansion at<br />

mid LMCA and good LMCA ostium coverage (mid panels). Final kissing balloon (right panel) allows PCI optimization.<br />

Fig. 4 e Final result. Excellent final angiographic (panel A) and IVUS (panel B) result with good LMCA stent expansion.<br />

New techniques and tools have allowed interventional<br />

cardiologists to tackle more and more complex lesions, but PCI<br />

to LMCA CTO represents a very rare situation. To the best of our<br />

knowledge, only 2 cases have been reported in the literature so<br />

far. 5,6 In order to warrant proper stent sizing and coverage of all<br />

diseased segments, IVUS generally plays a relevant role in the<br />

context of LMCA PCI. In the present case of a LMCA CTO, IVUS<br />

guidance revealed essential, as contrast opacification through<br />

the channel opened by multiple predilations did not clarify the<br />

real caliber and length of the vessel. Conversely, IVUS pullback<br />

allowed to visualize a concentric tubular disease from proximal<br />

Cx to ostial LMCA and guided the choice of the size and the<br />

length of the stent. Soon after stent deployment, IVUS run from<br />

LAD showed minor plaque shift to proximal LAD and ostial<br />

LMCA coverage but suboptimal stent expansion throughout all<br />

its length. PCI was then optimized with kissing balloon for LAD<br />

and Cx ostia and distal LMCA and with a large NC balloon for<br />

mid-to-proximal LMCA stent. Optimal stent apposition was<br />

confirmed by final IVUS run.<br />

4. Conclusions<br />

This case demonstrates CTO of the LMCA, although a rare<br />

finding, is compatible with life and can even present without<br />

typical angina in a non-diabetic patient. PCI of LMCA CTO is<br />

feasible in experienced hands and, owing to the reestablishment<br />

of normal anatomy, provides a more physiological<br />

revascularization than a surgical approach. In these procedures,<br />

IVUS guidance reveals of crucial importance to<br />

understand the anatomy and to guide stent sizing and<br />

placement.


indian heart journal 65 (<strong>2013</strong>) 194e197 197<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Greenspan M, Iskandrian AS, Segal BL, Kimbiris D, Bemis CE.<br />

Complete occlusion of the left main coronary artery. Am <strong>Heart</strong><br />

J. 1979;98:83e86.<br />

2. Zimmern SH, Rogers WJ, Bream PR, et al. Total occlusion of the<br />

left main coronary artery: the coronary artery surgery study<br />

(CASS) experience. Am J Cardiol. 1982;49:2003e2010.<br />

3. Takagi H, Kawai N, Umemoto T. Stenting versus coronary<br />

artery bypass grafting for unprotected left main coronary<br />

artery disease: a meta-analysis of comparative studies. J Thorac<br />

Cardiovasc Surg. 2009;137:54e57.<br />

4. Morice MC, Serruys PW, Kappetein AP, et al. Outcomes in patients<br />

with de novo left main disease treated with either percutaneous<br />

coronary intervention using paclitaxel-eluting stents or coronary<br />

artery bypass graft treatment in the Synergy between<br />

Percutaneous Coronary Intervention with TAXUS and Cardiac<br />

Surgery (SYNTAX) trial. Circulation. 2010;121:2645e2653.<br />

5. Koster NK, White M. Chronic effort-induced angina as<br />

presentation of a totally occluded left main coronary artery: a<br />

case report and review. Angiology. 2009;60:382e384.<br />

6. Secco GG, <strong>Mar</strong>ino PN, Venegoni L, De Luca G. Percutaneous<br />

revascularization of chronic total occlusion of the left main<br />

coronary artery. Rev Esp Cardiol. 2011;64:431e433.<br />

Book Review<br />

Pediatric Cardiac Intensive Care. Pre and Postoperative<br />

Guidelines, Manoj Luthra. Elsevier A Division of Reed Elsevier<br />

India Pvt Ltd, 3, Tolstoy <strong>Mar</strong>g, New Delhi, India, (2012).<br />

Pages: 314, Price: INR 475/-<br />

Pediatric Cardiac Intensive Care has seen a tremendous<br />

progress over the last few decades in our country. The centers<br />

providing this specialized care have increased steadily with<br />

newer centers being added every year across non-metro cities<br />

too. This has created a requirement of good pediatric cardiac<br />

intensive care services which most often is delivered by the<br />

team of pediatric cardiac surgeon, pediatric cardiac anesthetist,<br />

pediatric cardiologist, intensivist, pediatrician, registrars<br />

and nurses at various levels. With this scenario, there is an<br />

urgent need for a simple yet informative and concise handbook<br />

to enable provision of quality care to some of the sickest<br />

of children with cardiac problems across their peri-operative<br />

and post-operative period.<br />

‘The Manual of Pediatric cardiac intensive care e Pre and<br />

postoperative guidelines’ by Dr Manoj Luthra, an eminent<br />

pediatric cardiac surgeon of our country, is an excellent<br />

handbook which should enable the team of pediatric cardiac<br />

intensive care providers to handle most of the situations<br />

arising in the unit. It has chapters on almost all the relevant<br />

emergencies such as congestive cardiac failure, arrhythmias,<br />

hypertension, pulmonary hypertension, post-operative respiratory<br />

complications, ARDS, ventilator associated pneumonia,<br />

sepsis, seizures, acute kidney injury and<br />

coagulopathies. Besides these emergencies, many basic<br />

physiology topics such as fluid and electrolytes, ABG analysis,<br />

hemodynamic monitoring, parenteral and enteral nutrition<br />

have been discussed very succinctly .It also contains valuable<br />

chapters on important drugs used in the PCCU and has a well<br />

compiled set of appendices at the end. The book is handy, is<br />

well illustrated and the author has kept the language very<br />

simple so that it can be used by the different levels of child<br />

care providers. The book is likely to be useful to anyone<br />

involved in looking after the sick child in the pediatric cardiac<br />

care unit.<br />

It is a difficult task to convert volumes of information<br />

available on pediatric cardiac intensive care to a few hundred<br />

pages. However, the author has created an excellent handbook<br />

on the subject and reflects decades of experience in<br />

practical day to day management in the PCCU and toward<br />

which the book shall go a long way in fulfilling the requirements<br />

of the pediatric cardiac care provider.<br />

B.M. John*, Amit Devgan<br />

Associate Professor, Department of Pediatrics, AFMC,<br />

Sholapur Road, Pune 411040, India<br />

*Corresponding author. Tel.: þ91 09372326660.<br />

E-mail address: drbmj1972@yahoo.com (B.M. John)


indian heart journal 65 (<strong>2013</strong>) 147e151<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Fractional Flow Reserve: Intracoronary versus intravenous<br />

adenosine induced maximal coronary hyperemia<br />

P.S. Sandhu a , Upendra Kaul a,b, *, R.K. Gupta a , Tapan Ghose a<br />

a Fortis Escorts <strong>Heart</strong> Institute and Research Center, Okhla Road, New Delhi, India<br />

b Executive Director and Dean, Fortis Flt. Lt Rajen Dhall Hospital & Fortis Escorts <strong>Heart</strong> Institute and Research Center,<br />

Okhla Road, New Delhi 1100, India<br />

article info<br />

Article history:<br />

Received 8 October 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 24 February <strong>2013</strong><br />

Keywords:<br />

Fractional Flow Reserve<br />

Adenosine<br />

Maximal hyperemia<br />

abstract<br />

Background: Fractional Flow Reserve (FFR), a measure of coronary stenosis severity is based<br />

on the achievement of maximal hyperemia of coronary microcirculation. The most widely<br />

used pharmacological agent is adenosine which is administered either by intra coronary or<br />

intra venous routes. IV route is time consuming, has more side effects and expensive. This<br />

study is undertaken to compare the two routes of administration.<br />

Methods: FFR was assessed in 50 patients with 56 intermediate focal lesions using both IV<br />

and intracoronary (IC) adenosine. FFR was calculated as the ratio of the distal coronary<br />

pressure to the aortic pressure at maximal hyperemia.<br />

Results: A total of 25 left anterior descending, 8 right, 21 circumflex, and 2 left main coronary<br />

arteries were evaluated. The mean percent stenosis was 63.91 13.13 SD and, the<br />

mean FFR was 0.831 0.0738 SD for IV and 0.832 0.0707 SD for IC adenosine. There was a<br />

strong and linear correlation between 2 sets of observations with IV dose and IC adenosine<br />

dose (R ¼ 0.964, y ¼ 0.065 þ 0.923x; p < 0.001) (y ¼ IV dose, x ¼ IC dose). The agreement<br />

between the two sets of measurements was also high, with a mean difference of:<br />

0.001 0.0197. The changes in heart rate and blood pressure were significantly higher in IV<br />

adenosine group. Different incremental doses were well tolerated, with fewer systemic<br />

adverse events with IC adenosine. Transient AV blocks were observed with both IV and IC<br />

adenosine.<br />

Conclusions: This study suggests that IC adenosine is equivalent to IV infusion for the<br />

determination of FFR. The administration of IC adenosine is easy to use, cost effective, safe<br />

and associated with fewer systemic events.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

1. Introduction<br />

The temporal and spatial resolution of coronary angiography<br />

(CAG) have meant that despite major advances in noninvasive<br />

cardiac imaging, CAG continues to remain the gold standard<br />

for the diagnosis and management of coronary artery disease.<br />

However, the 2-dimensional silhouette image provided by<br />

CAG has well recognized limitations; it does not accurately<br />

* Corresponding author.<br />

E-mail address: kaul.upendra@gmail.com (U. Kaul).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.006


148<br />

indian heart journal 65 (<strong>2013</strong>) 147e151<br />

represent the true complexity of the coronary luminal<br />

morphology, and gives no indication of the functional influence<br />

of luminal changes on coronary blood flow. These limitations<br />

are more pronounced in angiographically intermediate<br />

stenosis (50e90%) and in patients in whom there is a clear<br />

discrepancy between the clinical picture and angiographic<br />

findings. 1e4<br />

The measurement of Fractional Flow Reserve (FFR) is used<br />

to determine the hemodynamic significance of epicardial<br />

coronary stenosis detected at CAG. 3 For an accurate calculation<br />

of FFR, the principle is to achieve a maximal hyperemia to<br />

minimize the contribution of microvascular resistance. 5 With<br />

suboptimal levels of coronary hyperemia, FFR will be artificially<br />

high, resulting in a potentially significant underestimation<br />

of the functional severity of the coronary stenosis.<br />

This physiologic method of assessing the severity of coronary<br />

lesions has become a very acceptable simple method utilized<br />

in a large numbers of cardiac catheterization laboratories<br />

world over after the results of FAME study were published. 4,5<br />

The method is being utilized in our country also increasingly.<br />

We have previously demonstrated its utility and cost<br />

saving advantage in our set. 6<br />

Although adenosine invariably intravenously (IV), has been<br />

recommended for FFR measurements, intracoronary (IC)<br />

administration of adenosine constitutes a valuable alternative<br />

in everyday practice. 4e9 Compared with the IC route, IV<br />

adenosine administration requires relatively large doses, and<br />

it is associated with more systemic adverse effects and<br />

costs. 10,11 However, several observations cast some doubts<br />

about the ability of the IC adenosine boluses to achieve<br />

maximal hyperemia in all patients. 11,12<br />

The aim of this study was to compare IV versus IC adenosine<br />

for calculating an accurate FFR at maximal hyperemia by<br />

two methods. The FFR was calculated by both methods in our<br />

setting at maximum hyperemia.<br />

of the procedure (5000 units). The heart rate and arterial<br />

pressure were continuously monitored throughout the procedure.<br />

After CAG, a 0.014-inch pressure-recording guidewire<br />

(PressureWire Certus, St. Jude Medical, USA) was introduced<br />

through a guiding catheter into the coronary artery. Arterial<br />

pressure wave damping or variation of the measured coronary<br />

guide pressure was avoided. The guidewire was externally<br />

calibrated and then advanced to the distal tip of the catheter. 14<br />

At this position, it was verified that both the catheter and the<br />

pressure wire recorded equal pressures. The pressure wire<br />

was subsequently advanced into the coronary artery with the<br />

pressure sensor placed beyond the lesion site. Distal coronary<br />

and aortic pressures were measured at baseline and at<br />

maximal hyperemia. Pressure signals were continuously<br />

recorded and a beat-to-beat analysis of mean pressure was<br />

performed automatically (RadiAnalyzer Xpress, St. Jude<br />

Medical, USA).<br />

2.3. Pharmacologic protocol<br />

All patients first received multiple IC adenosine boluses,<br />

which were then followed by IV adenosine. Incremental doses<br />

of IC adenosine (60, 100, and 120 mg for both coronary arteries)<br />

were administered. Each bolus was followed by a flush with<br />

5 ml saline. Subsequent doses were given after pressure<br />

curves returned to baseline values. Thereafter, adenosine<br />

infusion via a large systemic vein at incremental doses of 140,<br />

160, 180 mg/kg/min was administered until a steady-state hyperemia<br />

was achieved for a minimal duration of 1 min 15 The<br />

electrocardiogram was simultaneously recorded.<br />

2.4. Calculations of pressure-derived FFR<br />

FFR is defined as the ratio of the hyperemic flow in a stenotic<br />

artery to the hyperemic flow in the same artery in the<br />

2. Methods<br />

2.1. Study population<br />

FFR was assessed in a total of 50 patients enrolled prospectively.<br />

The study population consisted of 36 males and 14 females<br />

with a mean age of 62 8 years. Most patients had<br />

normal left ventricular function, and only focal or short<br />

segment of coronary artery stenosis ranging from 50e90%, as<br />

assessed by QCA, were analyzed. In all patients, FFR was<br />

determined for a target coronary lesion by both IV and IC<br />

adenosine. All patients were symptomatic with angina or<br />

angina equivalent and had been referred for a diagnostic or<br />

interventional cardiac catheterization. Exclusion criteria<br />

included culprit vessel in acute coronary syndrome, acute<br />

myocardial infarction, and atrioventricular conduction abnormalities<br />

in the electrocardiogram. All patients gave<br />

informed consent to participate in the study.<br />

2.2. Study protocol<br />

Coronary angiography (CAG) was performed from femoral or<br />

radial approach. Heparin was administered at the beginning<br />

Table 1 e Baseline demographic data of the study<br />

population.<br />

Study cohort (50 patients, 56 lesions)<br />

Age (yr.) 62 8<br />

Male/Female 36/14<br />

Risk factors<br />

Hypertension 18 (36%)<br />

Diabetes 21 (42%)<br />

Smoking 10 (20%)<br />

Dyslipidemia 15 (30%)<br />

Old MI 5 (10%)<br />

Angiographic parameters<br />

Single vessel disease 13 (26%)<br />

Double vessel disease 26 (52%)<br />

Triple vessel disease 11 (22%)<br />

Percent stenosis (%) 63.9 13.1<br />

Target vessel<br />

LAD 25 (44.6%)<br />

LCX 21 (37.5%)<br />

RCA 8 (14.3%)<br />

LM 2 (3.6%)<br />

Ejection fraction (%) 55 5%<br />

LAD, Left anterior descending artery; RCA, right coronary artery;<br />

LCx, left circumflex artery; LM, left main coronary artery.


indian heart journal 65 (<strong>2013</strong>) 147e151 149<br />

Fig. 1 e Linear regression analysis of FFR measurements performed with intracoronary (IC) (dependent variable) and<br />

intravenous (IV) 9 independent variable adenosine.<br />

hypothetical case when there is no stenosis present. 16 FFR<br />

expresses maximum hyperemic blood flow in a stenotic vessel<br />

as a fraction of normal maximal blood flow in that vessel. FFR is<br />

calculated from intracoronary and aortic pressure measurements<br />

obtained during maximal hyperemia by the following<br />

equation: FFR ¼ P d P v /P a P v , or FFRyP d /P a (if P v is negligible),<br />

where P a is the mean proximal coronary pressure, P d is the<br />

mean distal coronary pressure, and P v is the mean central<br />

venous pressure. FFR measurement was done with IC adenosine<br />

at incremental doses. The values were taken at maximum<br />

hyperemia. This was compared with IV adenosine at sequentially<br />

increasing doses till maximum hyperemia was produced.<br />

2.5. Statistical analysis<br />

Data are presented as mean SD. Student paired t test was<br />

used to compare FFR values after different hyperemic<br />

responses for IV and IC adenosine. Linear regression was<br />

calculated for FFR data derived from both hyperemic stimuli,<br />

and nonlinear regression was used for the relation of percent<br />

stenosis to the FFR. The mean SD of the signed differences<br />

between measurements of FFR with intravenous and intracoronary<br />

adenosine was used as an index of agreement between<br />

measurements. Results were considered statistically<br />

significant at p 0.05.<br />

3. Results<br />

3.1. Patient characteristics<br />

A total of 50 patients with 56 lesions were included in the<br />

analysis. Baseline demographic data is provided in Table 1.<br />

Procedural success was 100% for crossing the target lesion<br />

Fig. 2 e BlandeAltman agreement between 2 sets of measurements. Difference between measurements with intracoronary<br />

(IC) and intravenous (IV) adenosine plotted against mean.


150<br />

indian heart journal 65 (<strong>2013</strong>) 147e151<br />

Table 2 e Hemodynamic data for intravenous and intracoronary adenosine.<br />

IC adenosine<br />

(mean SD)<br />

IV adenosine<br />

(mean SD)<br />

p value<br />

DHR (beats/min) 0.8 3.4 5 5.6


indian heart journal 65 (<strong>2013</strong>) 147e151 151<br />

5. Conclusions<br />

This study suggests that IC adenosine is equivalent to IV<br />

infusion for achievement of maximal hyperemia and the<br />

determination of FFR in short segment or focal lesions. The<br />

administration of IC adenosine is easy to use, cost effective,<br />

safe and associated with fewer systemic events.<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Kern JM, De Bruyne B. From research to clinical practice:<br />

current role of intracoronary physiologically based decision<br />

making in the cardiac catheterization laboratory. JACC.<br />

1997;30:613e620.<br />

2. Kern JM. Coronary physiology revisited. Practical insights<br />

from the cardiac catheterization laboratory. Circulation.<br />

2000;101:1344e1351.<br />

3. Topol EJ, Nissen SE. Our preoccupation with coronary<br />

luminology: the dissociation between clinical and<br />

angiographic findings in ischemic heart disease. Circulation.<br />

1995;92:2333e2342.<br />

4. Tonino PA, De Bruyne B, Pijls NH, et al. FFR versus<br />

angiography for guiding PCI. NEJM. 2009;360:213e224.<br />

5. De Bruyne B, Pijls NH, Kalesan B, et al. FFR-guided PCI versus<br />

medical therapy in stable coronary disease. NEJM.<br />

2012;367:991e1001.<br />

6. Kaul U, Rastogi V, Seth A, et al. Role of fractional flow reserve<br />

(FFR) in decision making during multivessel PCI and the cost<br />

effectiveness. Our experience. IHJ. 2011;63:6.<br />

7. Jeremias A, Whitbourn RJ, Filardo SD, et al. Adequacy of IC vs<br />

IV adenosine-induced maximal coronary hyperemia for FFR<br />

measurements. AHJ. 2000;140:651e667.<br />

8. Casella G, Leibig M, Schiele TM, et al. Are high doses of IC<br />

adenosine an alternative to standard intravenous adenosine<br />

for the assessment of FFR? AHJ. 2004;148:590e595.<br />

9. Leone AM, Porto I, de Caterina AR, et al. IC vs IC sodium<br />

nitroprusside vs IV adenosine: the NASCI study. JACC Intv.<br />

2012;5:402e408.<br />

10. Wilson RF, Wyche K, Christensen BV, et al. Effects of<br />

adenosine on human coronary circulation. Circulation.<br />

1990;82:1595e1606.<br />

11. Kern MJ, Deligonul U, Tatineni S, et al. IV adenosine:<br />

continuous infusion and low dose bolus administration for<br />

determination of coronary vasodilator reserve in patients<br />

with and without coronary artery disease. JACC.<br />

1991;18:718e729.<br />

12. Jeremias A, Filardo SD, Whitbourn RJ, et al. Effect of IV and IC<br />

adenosine 5’- triphosphate as compared with adenosine on<br />

coronary flow and pressure dynamics. Circulation.<br />

2000;101:318e323.<br />

13. de Bruyne B, Bartunek J, Sys SU, et al. Simultaneous<br />

coronary pressure and flow velocity measurements in<br />

humans: feasibility, reproducibility, and hemodynamic<br />

dependence of coronary flow velocity reserve, hyperemic<br />

flow versus pressure slope index, and FFR. Circulation.<br />

1996;94:1842e1849.<br />

14. Pijls NH, van Gelder B, van der Voort P, et al. FFR: a useful<br />

index to evaluate the influence of an epicardial coronary<br />

stenosis on myocardial blood flow. Circulation.<br />

1995;92:3183e3193.<br />

15. Wilson RF, Wyche K, Christensen BV, et al. Effects of<br />

adenosine on human coronary arterial circulation. Circulation.<br />

1990;82:1595e1606.<br />

16. Pijls NH, van Son JA, Kirkeeide RL, et al. Experimental basis of<br />

determining maximum coronary, myocardial, and collateral<br />

blood flow by pressure measurements for assessing<br />

functional stenosis severity before and after percutaneous<br />

transluminal coronary angioplasty. Circulation.<br />

1993;87:1354e1367.<br />

17. Pijls NHJ, Klauss V, Siebert U, et al. Coronary pressure<br />

measurements after stenting predicts adverse events at<br />

follow-up: a multicenter registry. Circulation.<br />

2002;105:2950e2954.


indian heart journal 65 (<strong>2013</strong>) 172e179<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Fragmented narrow QRS complex: Predictor of left ventricular<br />

dyssynchrony in non-ischemic dilated cardiomyopathy<br />

Jamal Yusuf a , Devendra Kumar Agrawal a,b, *, Saibal Mukhopadhyay a , Vimal Mehta a ,<br />

Vijay Trehan a , Sanjay Tyagi a<br />

a Department of Cardiology, G.B. Pant Hospital, New Delhi, India<br />

b Senior Resident, Department of Cardiology, Maulana Azad Medical College and G.B. Pant Hospital, Jawaharlal Nehru <strong>Mar</strong>g,<br />

New Delhi 110002, India<br />

article info<br />

Article history:<br />

Received 20 September 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 4 <strong>Mar</strong>ch <strong>2013</strong><br />

Keywords:<br />

Cardiomyopathy<br />

Intraventricular dyssynchrony<br />

Fragmented QRS complex<br />

Tissue Doppler imaging<br />

abstract<br />

Background: Cardiac resynchronization therapy is an important therapeutic modality in<br />

drug refractory symptomatic patients of heart failure with wide QRS (120 ms) on electrocardiogram.<br />

However, wide QRS (considered as a marker of electrical dyssynchrony)<br />

occurs in only 30% of heart failure patients, making majority of drug refractory heart failure<br />

patients ineligible for resynchronization therapy. Significant numbers of patients with<br />

narrow QRS have echocardiographic evidence of left ventricular dyssynchrony. However,<br />

there is sparse data about additional features on the surface ECG which can predict<br />

intraventricular dyssynchrony. This study was undertaken to assess the utility of fragmented<br />

narrow QRS complex to predict significant intraventricular dyssynchrony in<br />

symptomatic patients of non-ischemic dilated cardiomyopathy.<br />

Method: 100 symptomatic patients of non-ischemic dilated cardiomyopathy with narrow<br />

QRS complexes (50 each with fragmented and normal QRS) were recruited. Tissue Doppler<br />

imaging was used to assess intraventricular dyssynchrony as per ‘Yu index’.<br />

Results: 78% patients (n ¼ 39) in fQRS complex group and 14% (n ¼ 7) in normal QRS complex<br />

group had significant intraventricular dyssynchrony (c 2 ¼ 20.61; p < 0.000005). fQRS complexes<br />

had 84.78% sensitivity, 79.62% specificity, a positive predictive value of 78% and<br />

negative predictive value of 86% to detect intraventricular dyssynchrony. fQRS also had<br />

sensitivity and specificity of 93% and 90% respectively to localize the dyssynchronous<br />

segment.<br />

Conclusion: fQRS is a marker of electrical dyssynchrony, which results in significant intraventricular<br />

dyssynchrony in patients of non-ischemic dilated cardiomyopathy and a narrow<br />

QRS interval. fQRS localizes the dyssynchronous segment and might be useful in<br />

identifying patients who can benefit from cardiac resynchronization therapy.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

* Corresponding author. Department of Cardiology, Maulana Azad Medical College and G.B. Pant Hospital, Jawaharlal Nehru <strong>Mar</strong>g, New<br />

Delhi 110002, India.<br />

E-mail addresses: drdevendra_123@yahoo.co.in, devendra.aiims@gmail.com (D.K. Agrawal).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.007


indian heart journal 65 (<strong>2013</strong>) 172e179 173<br />

1. Introduction<br />

Cardiac resynchronization therapy (CRT) is a useful therapeutic<br />

strategy in drug refractory symptomatic patients of<br />

heart failure (HF) with wide QRS (120 ms) on electrocardiogram<br />

(ECG). 1 Though QRS duration 120 ms has been adopted<br />

as a marker of electrical dyssynchrony responsible for presence<br />

of left ventricular (LV) dyssynchrony and subsequent<br />

eligibility for CRT, studies suggest that QRS widening 120 ms<br />

occurs in only 30% of HF patients. 2,3 In patients of HF with<br />

normal QRS duration, LV dyssynchrony has been reported in<br />

20e50% patients. 4e7 However, there are not enough studies<br />

available analyzing additional features on the surface ECG that<br />

can be taken as marker of electrical dyssynchrony resulting in<br />

significant intraventricular dyssynchrony (IVD) in patients of<br />

HF with narrow QRS. Till date only two studies have reported<br />

that fQRS on the surface ECG is a predictor of significant IVD in<br />

patients of non-ischemic DCM. 8,9 We undertook this study to<br />

assess the sensitivity, specificity and positive predictive value<br />

(PPV) of fQRS complex on the surface ECG to detect significant<br />

IVD in symptomatic patients of non-ischemic DCM.<br />

2. Methods<br />

2.1. Patients selection<br />

This study was conducted in GB Pant Hospital, New Delhi, a<br />

tertiary care centre, located in the Northern part of India. The<br />

study protocol conforms to the ethical guidelines of the 1975<br />

Declaration of Helsinki as reflected in a priori approval by the<br />

institution’s human research committee.<br />

A total of 100 patients of chronic HF due to non-ischemic<br />

dilated cardiomyopathy (DCM), with LV ejection fraction of<br />

less than 35% and sinus rhythm having narrow QRS complexes<br />

(


174<br />

indian heart journal 65 (<strong>2013</strong>) 172e179<br />

presence of significant (50% stenosis in any epicardial coronary<br />

artery) coronary artery disease (CAD). Patients with<br />

organic valvular heart disease, history suggestive of CAD,<br />

atrial fibrillation, kidney failure, liver failure and on permanent<br />

pacemakers were excluded from the study. The study<br />

protocol was approved by the institutional review board.<br />

Written informed consent was obtained from all participants.<br />

2.2. ECG<br />

The resting 12-lead ECG (filter range, 0.05e100 Hz; A.C. filter,<br />

60 Hz, 25 mm/s, 10 mm/mV) was analyzed by two independent<br />

clinicians who were blinded to echocardiographic data.<br />

The fQRS included various RSR’ patterns and was defined by<br />

the presence of an additional R-wave (R 0 prime), notching in<br />

nadir of the S-wave, notching of R-wave, or the presence of<br />

more than one R prime (fragmentation) in two contiguous<br />

leads corresponding to a major myocardial segment as previously<br />

described by Das et al (Fig. 1). 10 The presence of fQRS in<br />

2 contiguous anterior leads (V1eV5) were assigned to anterior<br />

myocardial segments, in 2 contiguous lateral leads (I,<br />

aVL, and V5, V6) to the lateral myocardial segments, in 2<br />

contiguous inferior leads (II, III, and aVF) to the inferior<br />

myocardial segments and in both V1 and V2 only to the posterior<br />

myocardial segments.<br />

2.3. Echocardiography<br />

Standard echocardiography with Doppler studies was performed<br />

by using a commercially available system (Philips iE33<br />

system, Andover, Massachusetts, USA). LV dimensions were<br />

measured by two-dimensional guided M-mode echocardiography<br />

according to the guidelines of the American Society of<br />

Echocardiography. 11 LV volumes were determined by the use<br />

of biplane Simpson’s method of disks, and the ejection fraction<br />

was calculated with a formula calling for the subtraction<br />

of the end-systolic volume from the end-diastolic volume and<br />

the difference divided by the end-diastolic volume. The<br />

effective orifice area and the regurgitant volume of the functional<br />

mitral regurgitation jet (MR volume) were calculated by<br />

‘proximal iso-velocity surface area’ method. 12<br />

Tissue Doppler imaging (TDI) was performed in the apical<br />

views (four-chamber, two-chamber and long-axis) for the long<br />

axis motion of the left ventricle. Two-dimensional echocardiography<br />

with tissue Doppler color imaging was performed<br />

with a 2.5 MHz phase array transducer. Myocardial regional<br />

velocity curves were constructed from the digitized images as<br />

previously described 13 (Fig. 2B). In this way septal, anteroseptal,<br />

anterior, lateral, inferior and posterior segments<br />

Fig. 1 e Various morphologies of QRS on 12-lead ECG<br />

defined as fQRS in present study originally proposed by<br />

Das et al. 10<br />

were interrogated at both basal and middle levels. Aortic<br />

opening and closure were determined by sampling the flow<br />

through the LV outflow tract with the use of pulsed Doppler<br />

echocardiography. For the purpose of measurement, the<br />

beginning of the QRS complex was used as the reference<br />

point, from where the time to peak myocardial sustained<br />

systolic (T s ) and early diastolic (T e ) velocities were calculated.<br />

The higher peak in velocity was selected when double peaks<br />

were encountered within the aortic ejection period.<br />

To assess global LV function, the myocardial sustained<br />

systolic (s), early diastolic (e) and late diastolic (a) velocities<br />

from the basal septal and basal lateral segments were calculated.<br />

For the assessment of synchronicity, the standard deviations<br />

of T s (T s -SD) of all 12 LV segments were calculated.<br />

Significant systolic IVD was defined as T s -SD >32.6 ms proposed<br />

by Yu et al and popularly known as ‘Yu index’. 14 Fig. 2B<br />

depicts color coded TDI (post processing image) of a patient<br />

with the ECG shown in Fig. 2A, showing systolic dyssynchronous<br />

inferior wall segments.<br />

We also looked for the association of fQRS and the most<br />

delayed contracting LV segment on echocardiography. ‘Dyssynchronic<br />

segments’ were defined as those LV segments that<br />

contracted later than 100 ms following the earliest contracting<br />

LV segment (which had the earliest T s ) and ‘most delayed<br />

segment’ was the last contracting myocardial segment (which<br />

had the latest T s ).<br />

2.4. Statistical analysis<br />

Analysis was performed using a statistical software program<br />

(SPSS for windows, version 17.0, SPSS Inc. Chicago, IL, USA).<br />

Data has been presented as mean SD and compared using<br />

the paired student’s t test. ManneWhitney U test has been<br />

used when the distribution was not normal. Categorical data<br />

between two or more groups were compared by Pearson c 2<br />

test. P value of


indian heart journal 65 (<strong>2013</strong>) 172e179 175<br />

Fig. 2 e (A): 12-lead ECG showing fragmented narrow QRS complexes in inferior leads (B): Colour coded TDI (post processing<br />

image) of the same patient showing systolic dyssynchronous inferior wall segments.<br />

Table 2 e Comparison of time to peak myocardial sustained systolic velocity (T s ) (in ms) between patients with fQRS and<br />

without fQRS.<br />

Segment Fragmented QRS (n ¼ 50) Normal QRS (n ¼ 50) p value<br />

Basal septal 158.52 37.86 160.26 33.09 0.80<br />

Mid septal 156.24 37.56 160.80 33.75 0.52<br />

Basal lateral 177.28 49.88 164.88 34.25 0.15<br />

Mid lateral 182.18 49.23 164.16 32.79 0.03*<br />

Basal inferior 187.48 48.44 169.64 32.64 0.03*<br />

Mid inferior 188.18 47.05 169.08 29.24 0.01*<br />

Basal anterior 177 47.21 161.36 28.38 0.048*<br />

Mid anterior 183.24 50.14 162.18 29.55 0.01*<br />

Basal posterior 172.16 40.25 167.08 29.08 0.47<br />

Mid posterior 173.76 45.23 169.84 31.05 0.61<br />

Basal anteroseptal 170.04 37.11 162.94 26.94 0.27<br />

Mid anteroseptal 169.7 38.52 169.90 34.23 0.98<br />

T s -SD (Yu index) 35.64 12.79 20.45 11.17


176<br />

indian heart journal 65 (<strong>2013</strong>) 172e179<br />

significant difference in severity of LV dilatation, LV ejection<br />

fraction and mitral regurgitation between the two groups.<br />

However, there was significant difference in the QRS<br />

duration in patients with fQRS complexes as compared to<br />

those with normal QRS, though QRS duration in both the<br />

groups was 32.6 ms) was seen in 39 patients (78%) in<br />

fQRS complex group and 7 patients (14%) in normal QRS<br />

complex group (Pearson’s c 2 ¼ 20.61; p < 0.000005) [Table 2].<br />

The presence of fQRS complexes in the basal ECG was<br />

found to detect systolic IVD as defined by the ‘Yu index’ (T s -<br />

SD > 32.6 ms) with sensitivity of 84.78%, specificity of 79.62%,<br />

positive predictive value (PPV) of 78% and a negative predictive<br />

value (NPV) of 86% (Table 3).<br />

The 50 patients with fQRS complexes were sub-divided<br />

into two groups with either significant systolic dyssynchrony<br />

(group 1, n ¼ 39) or without dyssynchrony (group 2,<br />

n ¼ 11) on the basis of T s -SD greater than or lesser than<br />

32.6 ms. Table 4 shows the clinical, demographic and echocardiographic<br />

characteristics of patients in group 1 and 2. The<br />

demographic and clinical parameters between the two groups<br />

were not statistically different. The patients in group 1 had<br />

significantly longer isovolumetric relaxation time (IVRT) in<br />

comparison to patients in group 2 (105.23 20.36 vs.<br />

72.00 8.90 ms; p < 0.001). There was no significant difference<br />

in any other echocardiographic parameters between the two<br />

groups. Among 39 patients of group 1 with significant systolic<br />

dyssynchrony, 30 (76.92%) patients had ECG fragmentation in<br />

the maximal dyssynchronic segment and six (15.39%) patients<br />

had ECG fragmentation in one of the dyssynchronic segments.<br />

In three (7.69%) patients, fragmented segments and dyssynchronic<br />

segments did not correlate. Among the dyssynchronic<br />

patients, the sensitivity and specificity of a<br />

fragmented segment on ECG to detect the most dyssynchronic<br />

segment or one of the dyssynchronic segments were 93% and<br />

88%, respectively.<br />

Table 3 e Relationship between fragmented QRS and<br />

intraventricular systolic dyssynchrony.<br />

Group<br />

Significant<br />

systolic<br />

dyssynchrony<br />

present (n)<br />

No significant<br />

systolic<br />

dyssynchrony<br />

(n)<br />

Total (n)<br />

Fragmented QRS 39 (true positives) 11 (false positives) 50<br />

Normal QRS 7 (false negatives) 43 (true negatives) 50<br />

Total 46 54 100<br />

4. Discussion<br />

CRT is an established treatment option for patients with drug<br />

refractory heart failure. The predominant mechanism of benefit<br />

from CRT appears to be related to the presence of IVD due to<br />

electrical conduction delay and subsequent resynchronization<br />

after CRT. 15e19 The presence of significant IVD at baseline secondary<br />

to electrical dyssynchrony is therefore, mandatory for<br />

the response to CRT. QRS duration of 120 ms has been adopted<br />

as a cut-off point for electrical dyssynchrony resulting in LV<br />

systolic dyssynchrony in CRT trials 20,21 and subsequently by<br />

treatment guidelines. 1 However, studies suggest that QRS<br />

widening of 120 ms occurs in only 30% of HF patients making<br />

majority of HF patients ineligible for CRT. On the other hand,<br />

significant IVD by TDI has been reported in 36e51% 14,22 of patients<br />

with HF and QRS 150 ms, the ‘Yu index’ has a sensitivity and<br />

specificity of 100% and 78%, respectively to predict reverse<br />

remodeling, while in patients with QRS duration of<br />

120e150 ms the sensitivity is 83% and specificity is 86%. 30 So<br />

we decided to use ‘Yu index’ to assess mechanical dyssynchrony<br />

in patients with fQRS and drug refractory symptomatic<br />

non-ischemic dilated cardiomyopathy as it may also serve as<br />

a predictor of response to CRT in this subgroup of patients<br />

with narrow QRS. Yu et al 14 developed this twelve segment


indian heart journal 65 (<strong>2013</strong>) 172e179 177<br />

Table 4 e Characteristics of patients with fQRS with (group 1) and without (group 2) significant systolic dyssynchrony.<br />

Characteristic<br />

Group 1 (with systolic<br />

dyssynchrony) (n ¼ 39)<br />

Group 2 (no systolic<br />

dyssynchrony) (n ¼ 11)<br />

Chi square value p value<br />

Age (in years) 46.13 14.24 45.00 12.20 0.80<br />

Sex (M:F) 23:16 9:2 0.43 0.51<br />

NYHA class n (%) 2.74 0.09<br />

II 23 (59%) 4 (57%)<br />

III-IV 16 (41%) 7 (43%)<br />

<strong>Heart</strong> rate (BPM) 91.67 17.74 98.09 14.91 0.24<br />

PR interval (ms) 148.59 29.52 155.82 31.49 0.51<br />

QRS duration (ms) 99.08 13.32 100.64 12.62 0.72<br />

LVEF % (Simpson’s) 22.31 5.30 22.00 4.43 0.85<br />

EPSS (mm) 20.47 3.44 21.33 1.70 0.26<br />

MR regurgitant volume (ml) 31.61 13.45 30.66 22.41 0.90<br />

TR jet gradient (mm Hg) 27.09 9.40 31.10 7.46 0.18<br />

Mitral E/A 2.29 1.39 3.03 1.70 0.21<br />

IVRT (ms) 105.23 20.36 72.00 8.90


178<br />

indian heart journal 65 (<strong>2013</strong>) 172e179<br />

patients with non-ischemic DCM and a narrow QRS interval.<br />

fQRS on the surface ECG is also helpful for localizing the<br />

dyssynchronous segment. In future, studies are needed to<br />

assess the beneficial effects of CRT in drug refractory, symptomatic<br />

HF patients with narrow fragmented QRS complexes<br />

and significant IVD.<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Epstein AE, Di<strong>Mar</strong>co JP, Ellenbogen KA, et al. ACC/AHA/HRS<br />

2008 guidelines for device-based therapy of cardiac rhythm<br />

abnormalities: a report of the American College of Cardiology/<br />

American <strong>Heart</strong> Association Task Force on Practice<br />

Guidelines (Writing Committee to Revise the ACC/AHA/<br />

NASPE 2002 Guideline Update for Implantation of Cardiac<br />

Pacemakers and Antiarrhythmia Devices) developed in<br />

collaboration with the American Association for Thoracic<br />

Surgery and Society of Thoracic Surgeons. J Am Coll Cardiol.<br />

2008;51:e1ee62.<br />

2. Kashani A, Barold SS. Significance of QRS complex duration<br />

in patients with heart failure. J Am Coll Cardiol. 2005;46:<br />

2183e2192.<br />

3. Freudenberger R, Sikora JA, Fisher M, et al. Electrocardiogram<br />

and clinical characteristics of patients referred for cardiac<br />

transplantation: implications for pacing in heart failure. Clin<br />

Cardiol. 2004;27:151e153.<br />

4. Bleeker G, Schalij M, Molhoek S, et al. Frequency of left<br />

ventricular dyssynchrony in patients with heart failure and a<br />

narrow QRS complex. Am J Cardiol. 2005;95:140e142.<br />

5. Auricchio A, Yu CM. Beyond the measurement of QRS<br />

complex toward mechanical dysynchrony: cardiac<br />

resynchronization therapy in heart failure patients with a<br />

normal QRS duration. <strong>Heart</strong>. 2004;90:479e481.<br />

6. Dohi K, Suffoletto M, Murali S, et al. Benefit of cardiac<br />

resynchronization therapy to a patient with a narrow<br />

QRS complex and ventricular dyssynchrony identified by<br />

tissue synchronization imaging. Eur J Echocardiogr.<br />

2005;6:455e460.<br />

7. Yu CM, Chan YS, Zhang Q, et al. Benefits of cardiac<br />

resynchronization therapy for heart failure patients with<br />

narrow QRS complexes and coexisting systolic asynchrony<br />

by echocardiography. J Am Coll Cardiol. 2006;48:2251e2257.<br />

8. Tigen K, Karaahmet T, Gurel E, et al. The utility of fragmented<br />

QRS complexes to predict significant intraventricular<br />

dyssynchrony in nonischemic dilated cardiomyopathy<br />

patients with a narrow QRS interval. Can J Cardiol. 2009;25(9):<br />

517e522.<br />

9. Basaran Y, Tigen K, Karaahmet T, et al. Fragmented QRS<br />

Complex are associated with cardiac fibrosis and significant<br />

intraventricular systolic dyssynchrony in nonischemic<br />

dilated cardiomyopathy patients with a narrow QRS interval.<br />

Echocardiography. 2011;28(1):62e68.<br />

10. Das M, Khan B, Jacob S, et al. Significance of a fragmented<br />

QRS complex versus a Q wave in patients with coronary<br />

artery disease. Circulation. 2006;113:2495e2501.<br />

11. Schiller NB, Shah PM, Crawford M, et al. Recommendations<br />

for quantitation of the left ventricle by two-dimensional<br />

echocardiography: American Society of Echocardiography<br />

Committee on Standards, Subcommittee on Quantitation of<br />

the Two-Dimensional Echocardiograms. J Am Soc Echocardiogr.<br />

1989;2:358e367.<br />

12. Shiota T, Jones M, Teien DE, et al. Evaluation of mitral<br />

regurgitation using a digitally determined colour Doppler flow<br />

convergence ‘centerline’ acceleration method: studies in an<br />

animal model with quantified mitral regurgitation. Circulation.<br />

1994;89:2879e2886.<br />

13. Gorcsan 3rd J, Strum DP, Mandarino WA, et al. Quantitative<br />

assessment of alterations in regional left ventricular<br />

contractibility with color-coded tissue Doppler<br />

echocardiography. Comparison with sonomicrometry and<br />

pressure volume relations. Circulation. 1997;95:2423e2433.<br />

14. Yu CM, Lin H, Zhang Q, et al. High prevalence of left<br />

ventricular systolic and diastolic asynchrony in patients with<br />

congestive heart failure and normal QRS duration. <strong>Heart</strong>.<br />

2003;89:54e60.<br />

15. Bax JJ, <strong>Mar</strong>wick TH, Molhoek SG, et al. Left ventricular<br />

dyssynchrony predicts benefit of cardiac resynchronization<br />

therapy in patients with end-stage heart failure before<br />

pacemaker implantation. Am J Cardiol. 2003;92:1238e1240.<br />

16. Yu CM, Fung WH, Lin H, et al. Predictors of left ventricular<br />

reverse remodeling after cardiac resynchronization therapy<br />

for heart failure secondary to idiopathic dilated or ischemic<br />

cardiomyopathy. Am J Cardiol. 2003;91:684e688.<br />

17. Breithardt OA, Stellbrink C, Kramer AP, et al.<br />

Echocardiographic quantification of left ventricular<br />

asynchrony predicts an acute hemodynamic benefit of<br />

cardiac resynchronization therapy. J Am Coll Cardiol.<br />

2002;40:536e545.<br />

18. Bordachar P, Lafitte S, Reuter S, et al. Echocardiographic<br />

parameters of ventricular dyssynchrony validation in<br />

patients with heart failure using sequential biventricular<br />

pacing. J Am Coll Cardiol. 2007;44:2157e2165.<br />

19. Bax JJ, Bleeker GB, <strong>Mar</strong>wick TH, et al. Left ventricular<br />

dyssynchrony predicts response and prognosis after cardiac<br />

resynchronization therapy. J Am Coll Cardiol.<br />

2004;44:1834e1840.<br />

20. Cleland JG, Daubert JC, Erdmann E, et al. The effect of cardiac<br />

resynchronization on morbidity and mortality in heart<br />

failure. N Engl J Med. 2005;352:1539e1549.<br />

21. Bristow MR, Saxon LA, Boehmer J, et al. Cardiacresynchronization<br />

therapy with or without an implantable<br />

defibrillator in advanced chronic heart failure. N Engl J Med.<br />

2004;350:2140e2150.<br />

22. Haghjoo M, Bagherzadeh A, Fazelifar AF, et al. Prevalence of<br />

mechanical dyssynchrony in heart failure patients with<br />

different QRS durations. Pacing Clin Electrophysiol.<br />

2007;30:616e622.<br />

23. Bleeker GB, Holman ER, Steendijk P, et al. Cardiac<br />

resynchronization therapy in patients with a narrow QRS<br />

complex. J Am Coll Cardiol. 2006;48:2243e2250.<br />

24. Achilli A, Sassara M, Ficili S, et al. Long-term effectiveness of<br />

cardiac resynchronization therapy in patients with refractory<br />

heart failure and "narrow" QRS. J Am Coll Cardiol.<br />

2003;42:2117e2124.<br />

25. Takemoto Y, Hozumi T, Sugioka K, et al. Beta-blocker therapy<br />

induces ventricular resynchronization in dilated<br />

cardiomyopathy with narrow QRS complex. J Am Coll Cardiol.<br />

2007;49:778e783.<br />

26. <strong>Mar</strong>cassa C, Campini R, Verna E, et al. Assessment of cardiac<br />

asynchrony by radionuclide phase analysis: correlation with<br />

ventricular function in patients with narrow or prolonged<br />

QRS interval. Eur J <strong>Heart</strong> Fail. 2007;9:484e490. 39.<br />

27. Timonen P, Magga J, Risteli J, et al. Cytokines, interstitial<br />

collagen and ventricular remodelling in dilated<br />

cardiomyopathy. Int J Cardiol. 2008;124:293e300.<br />

28. <strong>Mar</strong>ijianowski MM, Teeling P, Mann J, et al. Dilated<br />

cardiomyopathy is associated with an increase in the type


indian heart journal 65 (<strong>2013</strong>) 172e179 179<br />

I/type III collagen ratio: a quantitative assessment. J Am Coll<br />

Cardiol. 1995;25:1263e1272.<br />

29. Gorcsan J, Abraham T, Agler DA, et al. Echocardiography for<br />

cardiac resynchronization therapy: recommendations for<br />

performance and reportingea report from the American Society<br />

of Echocardiography Dyssynchrony Writing Group endorsed by<br />

the <strong>Heart</strong> Rhythm Society. JAmSocEchocardiogr. 2008;21:191e213.<br />

30. Yu CM, Fung JW, Chan CK, et al. Comparison of efficacy of<br />

reverse remodeling and clinical improvement for relatively<br />

narrow and wide QRS complexes after cardiac<br />

resynchronization therapy for heart failure. J Cardiovasc<br />

Electrophysiol. 2004;15:1058e1065.<br />

31. Beshai JF, Grimm RA, Nagueh SF, et al. Cardiacresynchronization<br />

therapy in heart failure with narrow QRS<br />

complexes. N Engl J Med. 2007;357:2461e2471.<br />

32. Chung ES, Leon AR, Tavazzi L, et al. Results of the predictors<br />

of response to CRT (PROSPECT) trial. Circulation.<br />

2008;117:2608e2616.


indian heart journal 65 (<strong>2013</strong>) 187e190<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Review Article<br />

“On” or “Off” pump coronary artery bypass grafting e Is the<br />

last word out?<br />

O.P. Yadava*, Anirban Kundu<br />

Department of Cardiac Surgery, National <strong>Heart</strong> Institute, New Delhi-110065, India<br />

A glance at the history of the development of Coronary Artery<br />

Bypass Surgery (CABG) throws up the interesting finding that<br />

the first milestones were without cardiopulmonary bypass<br />

(CPB) support. Off-pump CABG (OPCAB), having predated Onpump<br />

surgery, has had a roller coaster ride for want of a clean,<br />

still and bloodless field, culminating in the introduction of<br />

CABG on CPB (On-pump) by Favaloro in 1967. This development<br />

profoundly “democratised” the CABG procedure in that<br />

now a broad number of surgeons could achieve better and<br />

reproducible results with considerably more optimum operating<br />

conditions. The initial enthusiasm for On-pump CABG<br />

gradually gave way to concerns regarding its safety, especially<br />

with regard to complications arising from CPB, and not CABG<br />

per se. Foremost of these relate to microembolic showering<br />

during manipulation of the aorta and neurocognitive<br />

dysfunction. In addition, CPB triggers a whole-body inflammatory<br />

response caused by contact activation of the complement<br />

cascade. This leads to multiple organ dysfunction<br />

affecting the kidneys, liver, lungs, brain and heart itself. 1<br />

Studies published over a decade and a half ago questioned<br />

the safety of On-pump CABG. The proportion of patients<br />

recovering without any complication was found to be only<br />

64.3%. 2 In addition, health insurance data and data from<br />

clinical studies showed that 10.2% did not leave the hospital<br />

within 14 days after the operation and 3.6% of the patients<br />

were discharged to a non-acute care facility. 3 These and other<br />

observations, pari passu with the development of mechanical<br />

and pharmacological organ stabilizers and intracoronary<br />

shunts, resurrected OPCAB in the early 1990s.<br />

As regards surgical technique, the actual suture anastomosis<br />

of the vessels follows the same technique both in Onand<br />

Off-pump surgery. The difference is that unlike On-pump<br />

surgery, where the heart is arrested by means of cardioplegia,<br />

in Off-pump surgery the area of interest is kept immobile with<br />

the help of organ stabilizers while the anastomosis is being<br />

performed. Pharmacologic agents like short-acting betablockers<br />

(e.g. Esmolol) or Adenosine are used in conjunction<br />

with these mechanical stabilizers to achieve the goal of a<br />

relatively motionless field. Exposure of the heart is aided by<br />

retraction devices like the “Starfish”, which lift up the apex<br />

from the pericardial cavity (Table 1). Retraction can also be<br />

achieved by means of folded sponges placed in the pericardial<br />

sac to lift the heart, or by pericardial hitching sutures. Distal<br />

coronary perfusion is maintained by means of flexible intracoronary<br />

shunts introduced into the coronary vessel prior to<br />

commencement of the anastomosis (Fig. 1). Although these<br />

devices are designed for one-time use, they may be reused<br />

after ETO sterilization. This assumes importance as far as<br />

cost-containment in a country like India is concerned, giving a<br />

further fillip to Off-pump surgery.<br />

Numerous large observational studies and small randomized<br />

controlled trials (RCT) have been published in the past 18<br />

years suggesting benefits from OPCAB. Among these are, a<br />

reduction in stroke, duration of post-operative ventilation,<br />

need for reoperation, bleeding, wound infection, renal failure,<br />

post-operative length of stay 4 and decreased atrial fibrillation<br />

and inotrope requirement. 5 A major study published in 2001<br />

(the Octopus trial), showing no major differences in cardiac,<br />

neurological and neuropsychological outcomes in patients<br />

operated On- and Off-pump was also a shot in the arm for Offpump<br />

surgery. 6 However, as with any new technique, initial<br />

enthusiasm was somewhat dampened by reports of incomplete<br />

revascularization using OPCAB. 7 This and other reports<br />

led to the first RCT comparing the two procedures, the Randomized<br />

On/Off bypass study (ROOBY) in 2009. At 1-year of<br />

follow-up, Off-pump patients had worse composite outcomes<br />

* Corresponding author.<br />

E-mail address: op_yadava@yahoo.com (O.P. Yadava).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.008


188<br />

indian heart journal 65 (<strong>2013</strong>) 187e190<br />

Table 1 e Commonly used stabilizer/retraction devices.<br />

Name of stabilization<br />

device<br />

Stabilization<br />

mechanism<br />

Pros<br />

Cons<br />

Medtronic-Utrecht Octopus Ò<br />

tissue stabilization system<br />

Suction Good stabilization for LAD Difficulty in LCx territory,<br />

cumbersome<br />

(discontinued)<br />

Octopus 1 Suction Good stabilization for LAD Bulky, cumbersome, difficulty in<br />

LCx, long arm causes less stability;<br />

(discontinued)<br />

Octopus 2 Suction Better stabilization, lesser pod<br />

height<br />

Octopus 3 Suction Malleable suction pods, better<br />

adaptation to heart contour<br />

Stiff pods may cause heart injury<br />

(discontinued)<br />

Increased horizontal motion; (in<br />

use)<br />

Octopus 4 Suction 360 arm movement (in use)<br />

Genzyme Elite stabilizer<br />

Compression of coronary<br />

between tapes<br />

No need for shunt<br />

Damage to coronary endothelium,<br />

possibility of catching back wall<br />

during anastomosis; (not widely<br />

used)<br />

Guidant system Suction High flexibility of arm Usable only on custom-made<br />

retractor (in use)<br />

Starfish positioning system Suction Allows for posterior vessel access May cause apical ischemia, injury<br />

(in use)<br />

of death, myocardial infarction (MI), graft patency and repeat<br />

revascularization. 8 The conversion rate from Off- to On-pump<br />

procedures was 12.4%, a figure 5 times that reported in the<br />

National Database of Thoracic Surgeons! This was bandied as<br />

glaring evidence of the participant practitioners’ inexperience.<br />

9 The authors had addressed the prickly issue of surgeon<br />

experience by doing a sensitivity analysis based on high volume<br />

(>50 pre-study cases) versus low volume operators, and<br />

had found no significant difference in outcomes. Nonetheless,<br />

many have questioned the figure of 50 cases as being insufficient<br />

to indicate surgeon experience, 10 whilst disregarding<br />

anesthesiologist experience.<br />

As if this was not enough, another nail in the coffin of<br />

OPCAB was driven by a recent Cochrane review meta-analysis<br />

11 from Copenhagen considering 10 low-bias trials<br />

(n ¼ 4950) of a total of 86 RCTs comparing Off-pump with Onpump<br />

surgery, which found 30% higher risk of all-cause<br />

mortality with Off- vs. On-pump CABG. Other adverse<br />

events like MI, stroke, renal insufficiency or repeat revascularization<br />

were similar despite a slightly lower number of<br />

distal anastomoses Off-pump versus On-pump. In fact the<br />

authors, Moller et al hazard to state, “findings suggest that<br />

funding sources matter, with the device industry-funded<br />

trials less apt to show harm from Off-pump CABG,” thereby<br />

suggesting that vested interests may be at play. But a major<br />

issue with the Cochrane review is that it included just one<br />

additional study to the meta-analysis by Afilalo et al (vide<br />

infra) which incidentally had showed clear superiority of<br />

Off-pump CABG. This additional study, ironically of Moller<br />

himself, had a staggering 25% mortality Off-pump, which<br />

obviously seriously skewed the results of the review to suggest<br />

harm with OPCAB.<br />

Fig. 1 e Diagrammatic representation of use of stabilizer & retraction/positioning devices and intracoronary shunts in<br />

Off-pump surgery. Reprinted from Verma S et al Off pump coronary artery bypass surgery. Fundamentals for the clinical<br />

cardiologist. Circulation. 2004;109:1206-1211.


indian heart journal 65 (<strong>2013</strong>) 187e190 189<br />

Angelini et al analyzed 2 RCTs comparing Off-pump and<br />

On-pump patients 6e8 years following surgery to assess graft<br />

patency, major adverse cardiac-related events (MACE), and<br />

health-related quality of life. The patient cohort followed up<br />

was from the Beating <strong>Heart</strong> Against Cardioplegic Arrest<br />

Studies (BHACAS) 1 and 2 trials. 12 Overall, 10.8% of grafts were<br />

occluded across both groups. Further, logistic regression<br />

analysis showed no evidence that grafts were more likely to be<br />

occluded in OPCAB than in On-pump patients. The differences<br />

in MACE-free survival and quality of Life indicators did not<br />

approach statistical significance. Although these findings<br />

greatly bolstered the case of the proponents of OPCAB surgery,<br />

some shortcomings pointed out were that the modality of<br />

assessment of graft patency was non-invasive (MDCTA) as<br />

against the gold standard of coronary angiography; also, the<br />

study was a single center study by a single surgical team, and<br />

hence could not necessarily be extrapolated to other surgeons<br />

and centers. Another concern with OPCAB is the lesser number<br />

of grafts put. But this has long been disproved by a review<br />

of 22 RCTs which showed only 0.2 fewer grafts being put in<br />

this group compared with On-pump. 13<br />

A meta-analysis published by Afilalo et al included<br />

all published and unpublished RCTs of Off-pump versus<br />

On-pump CABG from the MEDLINE, EMBASE and Cochrane<br />

databases. It took into consideration a total of 59 trials,<br />

encompassing 8961 patients. There was a significant 30%<br />

reduction in post-operative strokes with Off-pump surgery<br />

[risk ratio (RR) 0.70, 95% CI: 0.49e0.99]. There was no significant<br />

difference in mortality (RR: 0.90, 95% CI: 0.63e1.30) or MI<br />

(pooled RR: 0.89, 95% CI: 0.69e1.13). In the meta-regression<br />

analysis, the effect of OPCAB on all of the clinical outcomes<br />

was similar regardless of mean age, proportion of females in<br />

the trial, number of grafts per patient, and trial publication<br />

date. 13<br />

The most recent comparison study of the two techniques<br />

is the CABG Off or On pump revascularization study (CORO-<br />

NARY Study) published in <strong>Mar</strong>ch this year. 14 The trial randomized<br />

4752 patients undergoing CABG to Off-pump or<br />

On-pump groups. There was no significant difference seen<br />

in the primary end-points of death, MI, stroke, or new renal<br />

failure requiring dialysis at 30 days (Table 2).<br />

There were, however, some differences in secondary outcomes<br />

(Table 3), with the Off-pump group showing advantages<br />

of less bleeding, respiratory infections, and acute kidney<br />

injury, but this group also had fewer grafts performed and had<br />

more repeat revascularizations thereby mandating longer<br />

term follow-ups before a final verdict.<br />

Table 2 e CORONARY: primary outcomes.<br />

Endpoint<br />

Off-pump<br />

(%)<br />

On-pump<br />

(%)<br />

HR (95% CI)<br />

Primary composite 9.8 10.3 0.95 (0.79e1.14)<br />

end point<br />

Death 2.5 2.5 1.02 (0.71e1.46)<br />

MI 6.7 7.2 0.93 (0.75e1.15)<br />

Stroke 1.0 1.1 0.89 (0.51e1.54)<br />

New renal failure 1.2 1.1 1.04 (0.61e1.76)<br />

Table 3 e CORONARY: secondary outcomes.<br />

End point<br />

Off-pump<br />

(%)<br />

On-pump<br />

(%)<br />

HR (95% CI)<br />

Repeat revascularization 0.7 0.2 4.01 (1.34e12.0)<br />

Respiratory failure<br />

5.9 7.5 0.79 (0.63e0.98)<br />

or infection<br />

Acute kidney injury 28.0 32.1 0.87 (0.80e0.96)<br />

Blood transfusion 50.7 63.3 0.80 (0.75e0.85)<br />

Reoperation for<br />

perioperative bleeding<br />

1.4 2.4 0.61 (0.40e0.93)<br />

The investigators thus struck a middle path by recommending<br />

either of the two techniques based on patient factors,<br />

provided the surgeon was competent in both. For<br />

example, if a patient had renal dysfunction or a heavily<br />

calcified aorta, OPCAB would be preferred.<br />

Much like the proverbial see-saw, a 1-year follow-up<br />

angiographic study of the ROOBY cohort was reported this<br />

year by Hattler at the AHA Congress in Orlando. He revealed<br />

that Off-pump patients had a lower saphenous vein patency<br />

than On-pump, but a similar arterial graft patency rate,<br />

leading to less effective revascularization. Grover in an<br />

editorial tried to reconcile the difference between the ROOBY<br />

and CORONARY trial findings. 15 The latter involved only surgeons<br />

experienced in Off-pump surgery, whereas the ROOBY<br />

trial also had trainees as operating surgeons. On a conservative<br />

note however, he cautioned that any firm conclusions<br />

would have to await long term follow-up results.<br />

Training of surgeons in OPCAB techniques has been an<br />

issue as it requires a great deal of focus and enthusiasm, apart<br />

from safety concerns. However, even this issue has been laid<br />

to rest by a recent study from the Bristol <strong>Heart</strong> Institute 16 that<br />

Off-pump surgery is not only safe and reproducible, but the<br />

technique can be taught effectively and safely to trainees with<br />

clinical outcomes unrelated to either the level of supervision<br />

or the seniority of the trainees.<br />

The final frontier for the Off-pump coronary surgeon remains<br />

the patient with left main disease as well as those with<br />

severely depressed LV function. Here too, lower mortality and<br />

complication rates have been demonstrated. 17 OPCAB has<br />

been validated with results quite similar to the CORONARY<br />

trial in left main stem disease also. Murzi et al showed that<br />

OPCAB was associated with lower in-hospital mortality (0.5%<br />

vs. 2.9%; p ¼ 0.001) and morbidity in terms of stroke, renal<br />

dysfunction and pulmonary complications/infections. 18 But<br />

the biggest bugbear of OPCAB, viz. fewer grafts (2.7 0.7 vs.<br />

3 0.7; p ¼ 0.001) and lower rate of complete revascularization<br />

(88.3% vs. 92%; p ¼ 0.04) continue to remain. On multivariate<br />

analysis, CPB was confirmed to be an independent predictor of<br />

in-hospital mortality (OR 5.74; p ¼ 0.001). The countervailing<br />

pros and cons negating each other lead to similar survivals at<br />

1.5 and 10 years, a fact validated by Cheng et al. 19 OPCAB has<br />

also been shown to give superior results in patients with<br />

chronic kidney disease. 20<br />

The International Society for Minimally Invasive Cardiothoracic<br />

Surgery (ISMICS) recommendations state that the use<br />

of Off-pump bypass reduces perioperative morbidity, neurocognitive<br />

dysfunction and hospital length of stay and should<br />

be considered especially in high-risk patients, for example,


190<br />

indian heart journal 65 (<strong>2013</strong>) 187e190<br />

those with severe ascending aortic calcification, liver disease,<br />

renal insufficiency or other systemic processes that may be<br />

exacerbated by CPB, in order to reduce morbidity and<br />

mortality. 21<br />

A balanced approach has been advocated by the American<br />

<strong>Heart</strong> Association in their 2011 Guidelines. 22 Based on available<br />

data, the guidelines contend, both approaches are<br />

reasonable, with certain factors tilting the balance one way or<br />

the other. For example, the oft-quoted instance of a patient<br />

with a heavily diseased aorta being more amenable to Offpump<br />

surgery. The European Guidelines (2010) on the other<br />

hand make no mention of Off-pump surgery, stating instead<br />

that over 70% of bypass surgeries worldwide are conducted<br />

On-pump 23 (perhaps a diplomatic way of making an undiplomatic<br />

point!).<br />

As for our experience at the National <strong>Heart</strong> Institute in<br />

Delhi of over 5000 cases done Off-pump with a conversion rate<br />

of less than 1% and mortality of 1.6%, we can safely state that<br />

this is an effective technique, provided it is adopted and<br />

practiced in spirit as well as letter. The surgeon, and perforce<br />

the team, should be well conversant with the nuances of Offpump<br />

techniques, besides being motivated, focused and inclined,<br />

and not be a proponent just to join the bandwagon and<br />

be counted, whilst jeopardizing the patient’s interest. In the<br />

end, a doctor’s reputation lives through his/her patients, and<br />

this is no truer than for a cardiac surgeon. The best technique<br />

is that which works best for that particular patient, in the<br />

context of his clinical setting and his treating surgeon’s<br />

repertoire lending credence to our strong belief that it is the<br />

surgeon and not the technique, which is at the heart of the<br />

problem.<br />

references<br />

1. De Jaegere PP, Suyker WJL. Off pump coronary artery bypass<br />

surgery. <strong>Heart</strong>. 2002;88(3):313e318.<br />

2. Duhaylongsod F. Minimally invasive cardiac surgery defined.<br />

Arch Surg. 2000;135:296e301.<br />

3. Roach G, Kanchuger M, Mangano C, et al. Adverse cerebral<br />

outcomes after coronary bypass surgery. N Engl J Med.<br />

1996;335:1857e1863.<br />

4. Cleveland Jr JC, Shroyer AL, Chen AY, et al. Off-pump<br />

coronary artery bypass grafting decreases risk-adjusted<br />

mortality and morbidity. Ann Thorac Surg. 2001;72:1282e1288.<br />

5. Angelini GD, Taylor FC, Reeves BC, et al. Early and midterm<br />

outcome after off-pump and on-pump surgery in Beating<br />

<strong>Heart</strong> against Cardioplegic Arrest Studies (BHACAS 1 and 2): a<br />

pooled analysis of two randomised controlled trials. Lancet.<br />

2002;359:1194e1199.<br />

6. Van Dijk D, Nierich AP, Eefting FD, et al. The Octopus Study:<br />

rationale and design of two randomized trials on medical<br />

effectiveness, safety, and cost-effectiveness of bypass surgery<br />

on the beating heart. Control Clin Trials. 2000;21(6):595e609.<br />

7. Hannan EL, Wu C, Smith CR, et al. Off-pump versus on-pump<br />

coronary artery bypass graft surgery: differences in shortterm<br />

outcomes and in long-term mortality and need for<br />

subsequent revascularization. Circulation.<br />

2007;116:1145e1152.<br />

8. Shroyer AL, Grover FL, Hattler B, et al, for the Veterans Affairs<br />

Randomized On/Off Bypass (ROOBY) Study Group. On-pump<br />

versus off-pump coronary artery bypass surgery. N Engl J Med.<br />

2009;361:1827e1837.<br />

9. Puskas JD, Mack MJ, Smith CR. On-pump versus off-pump<br />

CABG. N Engl J Med. 2010;362:851e854.<br />

10. Kieser TM. On-pump versus off-pump CABG. N Engl J Med.<br />

2010;362:852e854.<br />

11. Møller CH, Penninga L, Wetterslev J, et al. Off-pump versus<br />

on-pump coronary artery bypass grafting for ischaemic heart<br />

disease. Cochrane Database Syst Rev 2012;(3):CD007224.<br />

12. Angelini GD, Culliford L, Smith DK, et al. Effects of on- and<br />

off-pump coronary artery surgery on graft patency, survival,<br />

and health-related quality of life: long-term follow-up of 2<br />

randomized controlled trials. J Thorac Cardiovasc Surg.<br />

2009;137(2):295e303.<br />

13. Afilalo A, Rasti M, Ohayon SM, et al. Off-pump vs. on-pump<br />

coronary artery bypass surgery: an updated meta-analysis<br />

and meta-regression of randomized trials. Eur <strong>Heart</strong> J.<br />

2012;33(10):1257e1267.<br />

14. Lamy A, Devereaux PJ, Prabhakaran D, et al. Off-pump or onpump<br />

coronary- artery bypass grafting at 30 days. N Engl J<br />

Med. 2012;366:1489e1497.<br />

15. Grover FL. Current status of off-pump coronary-artery bypass<br />

[Editorial]. N Engl J Med. <strong>Mar</strong>ch 26, 2012;366. Published in<br />

NEJM.org.<br />

16. Murzi M, Caputo M, Aresu G, et al. Training residents in offpump<br />

coronary artery bypass surgery: a 14 year experience.<br />

J Thorac Cardiovasc Surg. 2012;143(6):1247e1253.<br />

17. Caputti GM, Palma JH, Gaia DF, et al. Off-pump coronary<br />

artery bypass surgery in selected patients is superior to the<br />

conventional approach for patients with severely depressed<br />

left ventricular function. Clinics (Sao Paulo). 2011<br />

December;66(12):2049e2053.<br />

18. Murzi M, Caputo M, Aresu G, et al. On-pump and off-pump<br />

coronary artery bypass grafting in patients with left main<br />

stem disease: a propensity score analysis. J Thorac Cardiovasc<br />

Surg. 2012;143(6):1382e1388.<br />

19. Cheng DC, Bainbridge D, <strong>Mar</strong>tin JE, et al, Evidence Based<br />

Perioperative Clinical Outcomes Research Group. Does offpump<br />

coronary artery bypass reduce mortality, morbidity,<br />

and resource utilization when compared with conventional<br />

coronary artery bypass? A meta-analysis of randomized<br />

trials. Anesthesiology. 2005;102:188e203.<br />

20. Schroeder ME, Chawla L, Zhao Y, et al. Effect of off-pump<br />

versus on-pump coronary artery bypass grafting in patients<br />

with chronic kidney disease. Crit Care. 2012;16(suppl 1):347.<br />

21. Puskas J, Cheng D, Knight J, et al. Off-pump versus<br />

conventional coronary artery bypass grafting: a metaanalysis<br />

and consensus statement from the 2004 ISMICS<br />

consensus conference. Innovations. 2005;1(1):3e27.<br />

22. ACCF/AHA Practice Guidelines. 2011 ACCF/AHA guideline<br />

for coronary artery bypass graft surgery. Circulation.<br />

2011;124:e659.<br />

23. The Task Force on Myocardial Revascularization of the<br />

European Society of Cardiology (ESC) and the European<br />

Association for Cardio-Thoracic Surgery (EACTS). Guidelines<br />

on myocardial revascularization. Eur <strong>Heart</strong> J.<br />

2010;31:2501e2555.


indian heart journal 65 (<strong>2013</strong>) 168e171<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Oral flecainide is effective in management of refractory<br />

tachycardia in infants<br />

Vikas Kohli*<br />

Pediatric Cardiology & Congenital Cardiac Surgery, Indraprastha Apollo Hospital, New Delhi, India<br />

article info<br />

Article history:<br />

Received 13 July 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 21 February <strong>2013</strong><br />

Keywords:<br />

Flecainide<br />

Tachycardia<br />

Digoxin<br />

abstract<br />

Background: Propranolol and digoxin have been used as first line drugs for treatment of<br />

supraventricular tachycardia (SVT) in infants. Flecainide and other drugs have been<br />

effective as a second line treatment for controlling refractory SVT.<br />

Material and methods: This is a prospective study without randomization and control. The<br />

inclusion criteria were: infants (12 months) with tachyarrhythmia who failed to respond<br />

to first line drugs. Patients having post-surgical arrhythmias were excluded from the study.<br />

Results: A total of 8 infants were treated with flecainide for refractory tachyarrhythmia’s.<br />

Diagnosis on electrocardiogram (ECG) was atrioventricular reentry tachycardia (AVRT) in 5,<br />

atrial ectopic tachycardia (AET) in 2, a combination of AVRT and atrioventricular nodal<br />

reentry tachycardia (AVNRT) in 1. All patients had failed trial of antiarrhythmic drugs prior<br />

to presentation: digoxin and propranolol in 7, amiodarone in 3, cardioversion in 1. Flecainide<br />

(80e130 mg/m 2 orally) resulted in termination of the tachycardia in all 8 patients.<br />

Acute pharmacological termination of arrhythmia occurred with oral flecainide loading in<br />

1 and temporarily with intravenous esmolol loading in 1 patient. Adjuvant therapy in form<br />

of propranolol was used in 5 and digoxin in 2. There were no side effects noted. Four episodes<br />

of recurrence were noted in 3 patients over 2 years, all of which responded to dose<br />

increase. Mean follow up time is 24.75 months.<br />

Conclusion: This small case series indicates that flecainide is an effective antiarrhythmic<br />

agent, free of side effects and when used orally is capable of terminating and controlling<br />

relatively resistant supraventricular tachycardia in children.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

1. Introduction<br />

Approximately, 60% of children with supraventricular tachycardia<br />

(SVT) develop their initial episode by 1 year of age. 1<br />

Most often it is managed with a single drug. Spontaneous<br />

resolution occurs in 50e80% of infants by 1 year of age. 2 In 10%<br />

of patients, first line drugs in the form of digoxin and/or betablocker<br />

may not be effective. These patients need second line<br />

drugs like flecainide, amiodarone, sotalol or propafenone.<br />

These drugs can be used either as monotherapy or in combination<br />

for control of tachycardia. 3e7 A few patients in this<br />

group may not be controlled with drugs and may rarely<br />

require radiofrequency ablation.<br />

Flecainide has been effective in controlling refractory SVT<br />

when used as a monotherapy or with other antiarrhythmics<br />

in children. 3,5e7 Flecainide has been effectively used<br />

* C-116 Sarita Vihar, New Delhi 110044, India. Tel.: þ91 9891362233; fax: þ91 11 26941746.<br />

E-mail addresses: vkohli_md@yahoo.com, pedsheart@gmail.com.<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.009


indian heart journal 65 (<strong>2013</strong>) 168e171 169<br />

intravenously for acute termination of arrhythmia, followed by<br />

oral maintenance for prevention of recurrence. Its use has been<br />

variable depending on a variety of factors. In a North American<br />

survey, less than 5% of patients received flecainide. 7 In India,<br />

intravenous flecainide is not available while the oral form is<br />

available only at limited locations. We present our experience<br />

with oral flecainide in acute and medium term control of<br />

arrhythmia in infants with SVT not controlled with first line<br />

drugs. Since intravenous flecainide is not available in India, we<br />

used oral flecainide. Sotalol and amiodarone would then have<br />

been used as the last line therapy, a strategy used by others too. 3<br />

2. Methods<br />

Data was prospectively collected, however, there was no<br />

randomization and there were no controls. The inclusion<br />

criteria were: infants (12 months) with a diagnosis of<br />

tachyarrhythmia with a failure to control SVT with first line<br />

drugs i.e. propranolol and digoxin in standard dosages and no<br />

history of cardiac surgery. The parameters recorded are<br />

shown in Tables 1 and 2. The QTc was calculated from surface<br />

electrocardiograms using Bazett’s formula.<br />

Echocardiogram was performed on all patients before<br />

initiation of therapy to assess anatomy as well as ventricular<br />

size and function. Twenty-four hour ambulatory ECG monitoring<br />

(Holter) was done prior to discharge and subsequently<br />

at 1 month follow up and then 3 monthly to assess efficacy of<br />

treatment in the first year of follow up following which at 6<br />

monthly intervals. This was done since follow up only on<br />

basis of history may not be reliable in infants. The objective of<br />

treatment was to get complete suppression of arrhythmia.<br />

Effective control of arrhythmia was defined as complete suppression<br />

of tachyarrhythmia on Holter monitoring.<br />

3. Results<br />

3.1. Patient profile<br />

A total of 8 patients met the inclusion criteria during the study<br />

period (January 2006 to December 2010). The mean age of<br />

patients was 5.1 months with the range of 2 monthse12<br />

months (Table 1). Seven infants had normal ventricular<br />

function by echocardiography and one had an ejection fraction<br />

of 40% of left ventricular function (patient no. 3), not low<br />

enough to contraindicate flecainide therapy. The decreased<br />

function was due to tachyarrhythmia associated cardiomyopathy<br />

which improved completely once the rate was<br />

controlled. At the time of presentation to our institution, all<br />

patients had a failed trial with more than one drug (4 had 2<br />

antiarrhythmics tried, 4 had 3 antiarrhythmics tried and in<br />

addition 1 had attempted cardioversion). All patients were in<br />

tachycardia when they presented to us. Three patients were<br />

on our follow up from neonatal period and had failure of first<br />

line medications when they were included in our study.<br />

3.2. Dosages and monitoring<br />

All patients were hemodynamically stable at presentation;<br />

oral maintenance dose was started in all except one (patient<br />

no. 4) where oral loading of flecainide was performed. Oral<br />

loading was done in the one patient with 120 mg/m 2 /day of<br />

flecainide divided in three doses over 12 h which resulted in<br />

termination of arrhythmia. No side effects were noted with<br />

oral loading. The median dose of flecainide at initiation of<br />

therapy was 80 mg/m 2 /day (range 70e100 mg/m 2 /day) and<br />

was given in two to three divided doses. Corrected QTc interval<br />

was noted prior to discharge and subsequently at follow<br />

up visits. The median dose required for the efficacy was<br />

100 mg/m 2 /day (range: 80e130 mg/m 2 /day). QTc was not<br />

allowed to increase beyond 0.46 ms and drug dose increase<br />

was stopped if QTc reached or exceeded 0.46 ms.<br />

Efficacy was defined as complete suppression of<br />

arrhythmia. Therapy was initiated in the hospital, and all infants<br />

were monitored as inpatients for 3e5 days prior to<br />

discharge. In view of the known interference of milk with<br />

flecainide absorption, we disallowed feeds half an hour before<br />

and after administration of flecainide. 8 Dose was adjusted for<br />

the body surface area as the baby grew. The dose was recalculated<br />

at monthly follow up visits and was rounded off<br />

to convenient dispensing dose (with the dilution of 10 mg/ml).<br />

3.3. Response to therapy<br />

All the patients reverted to normal sinus rhythm (8/8). Sinus<br />

rhythm was restored in less than 3 days in 2/8 patients and<br />

Table 1 e Study patient profiles.<br />

Patient Age (months) Cardiac anatomy ECG diagnosis Previous drugs used/<br />

cardioversion<br />

1 2 Normal AVRT & AVNRT D,P,A<br />

2 3 Ebstein’s AVRT D,E,A<br />

3 2 Normal AET D,P<br />

4 12 Normal AVRT D,P,A<br />

5 2 Normal AVRT D,P,CV<br />

6 12 Normal AVRT D,P<br />

7 6 Normal AVRT D,P, A<br />

8 2 Normal AET D,P<br />

Abbreviations: AVRT ¼ atrioventricular reentry tachycardia, AVNRT ¼ atrioventricular nodal reentry tachycardia, AET ¼ Atrial ectopic tachycardia,<br />

VT ¼ Ventricular tachycardia.<br />

D ¼ digoxin; P ¼ propranolol; E ¼ esmolol; A ¼ amiodarone; CV ¼ cardioversion.


170<br />

indian heart journal 65 (<strong>2013</strong>) 168e171<br />

Table 2 e Response to therapy.<br />

Patient Maximum dosage of<br />

flecainide used mg/m 2<br />

Time to complete<br />

control<br />

Additional<br />

drugs used<br />

Length<br />

of f/u (mo)<br />

Recurrences (months<br />

after therapy started)<br />

1 80 3 days P 48 1 (1 mo)<br />

2 120 3 days E b ,P,D,A a 36 2 (1 mo; 6 mo)<br />

3 100 1 day P,D 24 1 (3 mo)<br />

4 110 1 day e 36 0<br />

5 100 1 day P 12 0<br />

6 200 1 day P 24 0<br />

7 120 1 day P 6 0<br />

8 120 1 day P 12 0<br />

D ¼ digoxin; P ¼ propranolol; E ¼ esmolol; A ¼ amiodarone; mo ¼ months.<br />

a Starred indicates trial of this drug was given but stopped in few weeks.<br />

b Indicates use only on day 1.<br />

within 24 h in 6/8 patients. Holter recordings prior to discharge<br />

showed complete control of arrhythmia in 8/8 patients.<br />

Recurrences were noted in 3 patients, 1 month after the<br />

control in one and 3 months later in another, whereas in third<br />

patient there were 2 recurrences: the first one occurred after 1<br />

months of follow up and the second after 6 months. All the<br />

recurrences responded to dose increase, none requiring hospitalization.<br />

The median duration of therapy was 24 months<br />

(range: 12e48 months). One patient in the series is currently<br />

off all medications and has had no recurrence of arrhythmia<br />

(patient no. 1).<br />

3.4. Additional drugs<br />

Additional drugs were used in 7/8 patients. Esmolol was used<br />

in 1 for acute control of arrhythmia which was continued as<br />

propranolol; propranolol was used in 7/8 patients (in most as a<br />

continuation of therapy initiated prior to presentation). In 7/8<br />

patients, the desired control was achieved with flecainide and<br />

propranolol combination and in 1 with flecainide alone. In 1<br />

patient (patient no. 2 with Ebstein’s anomaly), amiodarone<br />

was tried for a short period during one of the recurrences<br />

without any effect and was discontinued. The recurrence was<br />

subsequently controlled by increasing the dosage of flecainide.<br />

Digoxin was used in 2/6 patients along with propranolol<br />

and flecainide.<br />

3.5. Follow-up<br />

All patients have been in sinus rhythm for a range of 6<br />

monthse48 months (mean 24.75 months). No pro-arrhythmic<br />

side effects were observed with flecainide. There was no<br />

episode of bradycardia or AV block. Echocardiograms<br />

demonstrated normal ventricular function in all infants while<br />

on flecainide. None of the patients had inadvertent prolongation<br />

of QTc (>0.48 s). Holter monitoring was performed at a<br />

6 monthly interval did not reveal recurrence of arrhythmia<br />

after the first 3 months in any of the patients except one.<br />

4. Discussion<br />

Supraventricular tachycardia in infants is usually controlled<br />

using digoxin or beta-blockers as first line therapy. 7 However,<br />

in about 10% of infants, second line therapy drugs like flecainide,<br />

propafenone, amiodarone or sotalol are needed as single<br />

agents or in combination. 3e7 Flecainide has been used as a<br />

second line agent, most often intravenously with a loading<br />

dose which would terminate the arrhythmia, followed by oral<br />

maintenance dosage. 9 Though Flecainide is a safe and effective<br />

drug its use is much less than anticipated especially in the<br />

North American continent mainly due to the results of the<br />

CAST trial in the late 80’s which showed a significant increase<br />

in mortality with Flecainide in myocardial infarction patients<br />

when it was used to prevent arrhythmias. 10 We have in this<br />

paper reported our experience with oral flecainide and noted<br />

good response in a small group of patients. This small study<br />

reinforces that oral flecainide provides a good intermediate<br />

option. There was only one patient in our series that did not<br />

respond to flecainide and went on to require amiodarone for<br />

control of his arrhythmia. Amiodarone has also been studied<br />

as a second line drug. 4,11 The intravenous amiodarone study<br />

group conducted a randomized double blinded trial of use of<br />

amiodarone in 61 children. The adverse event rate was 87% in<br />

this group. 11 Burri et al reported 23 infants with lifethreatening<br />

incessant tachycardia, predominantly supraventricular.<br />

4 Intravenous amiodarone was used as a single antiarrhythmic<br />

agent. In this study, amiodarone was effective in<br />

19/23 infants. Significant side effects were however noted:<br />

neurological side effects in one infant; liver enzyme elevation<br />

necessitating stopping amiodarone administration and sinus<br />

bradycardia requiring dose change in two patients. In<br />

contrast, our small group of patients using flecainide exhibited<br />

no side effects. A staged approach has recently been reported<br />

by Drago et al. 3 They tried a stepwise protocol on a<br />

study group of 55 infants. Initial treatment was with propafenone,<br />

flecainide or propranolol and only if not controlled<br />

with these was amiodarone used.<br />

Only about 30% patients required amiodarone alone or in<br />

combination. Others have tried Flecainide as first line therapy<br />

in newborns and it has worked successfully in 85% of the<br />

babies over a 24 month period. 12 Other reports include successful<br />

control of arrhythmia with a combination of amiodarone<br />

and flecainide or amiodarone and sotalol in this age<br />

group of patients. 5,6 In our study, we have used flecainide by<br />

the oral route which has been easy to administer and overcomes<br />

the problem of non-availability of intravenous flecainide<br />

in our part of the world. We have used propranolol and


indian heart journal 65 (<strong>2013</strong>) 168e171 171<br />

digoxin in addition in some patients, for a short duration as an<br />

overlap with flecainide. The limitations of this study include<br />

the small number of patients enrolled. A larger study would be<br />

required in order to more conclusively validate the use of oral<br />

flecainide. In addition, our patients, though presenting in<br />

tachycardia, were hemodynamically stable. This implies that<br />

there is an internal bias of non-critical patients in our subset.<br />

Non-availability of drug level monitoring has also been a<br />

limitation of this study. Esmolol and oral loading of flecainide<br />

were used when urgent control was required. The advantage<br />

offered by oral flecainide is the ease of administration and the<br />

degree of control of arrhythmia. Significantly, side effects<br />

were not an issue in this series and similar series reported by<br />

other authors using flecainide. 4e6<br />

5. Conclusions<br />

In this study, oral flecainide has been shown to be useful<br />

either alone or in combination as second line management in<br />

difficult to treat arrhythmia in infants less than 12 months<br />

age. Flecainide is safe and has shown good control of the<br />

arrhythmia even when given orally. It has also been safely<br />

administered in small children in this study over a long period<br />

of time. Apart from being useful for acute control, it is also<br />

effective in preventing the recurrence of arrhythmia in majority<br />

of the cases.<br />

Conflicts of interest<br />

The author has none to declare.<br />

references<br />

1. Tortoriello TA, Snyder CS, Smith EO, Fenrich Jr AL,<br />

Friedman RA, Kertesz NJ. Frequency of recurrence among<br />

infants with supraventricular tachycardia and comparison<br />

of recurrence rates among those with and without<br />

preexcitation and among those with and without response<br />

to digoxin and/or propranolol therapy. Am J Cardiol.<br />

2003;92(9):1045e1049.<br />

2. Garson Jr A, Gillette PC, McNamara DG. Supraventricular<br />

tachycardia in children: clinical features, response to<br />

treatment, and long-term follow-up in 217 patients. J Pediatr.<br />

1981;98(6):875e882.<br />

3. Drago F, Silvetti MS, De Santis A, et al. Paroxysmal<br />

reciprocating supraventricular tachycardia in infants:<br />

electrophysiologically guided medical treatment and longterm<br />

evolution of the re-entry circuit. Europace.<br />

2008;10:629e635.<br />

4. Burri S, Hug MI, Bauersfeld U. Efficacy and safety of<br />

intravenous amiodarone for incessant tachycardias in<br />

infants. Eur J Pediatr. 2003;162(12):880e884.<br />

5. Price JF, Kertesz NJ, Snyder CS, Friedman RA, Fenrich AL.<br />

Flecainide and sotalol: a new combination therapy for<br />

refractory supraventricular tachycardia in children


indian heart journal 65 (<strong>2013</strong>) 127e131<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Effects of a yoga intervention on lipid profiles of diabetes<br />

patients with dyslipidemia<br />

Nisha Shantakumari a, *, Shiefa Sequeira b , Rasha El deeb a<br />

a Department of Physiology, Gulf Medical University, Ajman, P.O. Box 4184, UAE<br />

b Fetal Medicine Genetic Center, Dubai Healthcare City, UAE<br />

article info<br />

Article history:<br />

Received 18 June 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 28 February <strong>2013</strong><br />

Keywords:<br />

Yoga<br />

Cholesterol<br />

Triglycerides<br />

Lipoproteins<br />

abstract<br />

Objective: The present study was conducted to assess the effectiveness of yoga in the<br />

management of dyslipidemia in patients of type 2 diabetes mellitus.<br />

Methods: This randomized parallel study was carried out in Medical College Trivandrum,<br />

Kerala, India. Hundred type 2 diabetics with dyslipidemia were randomized into control<br />

and yoga groups. The control group was prescribed oral hypoglycemic drugs. The yoga<br />

group practiced yoga daily for 1 h duration along with oral hypoglycemic drugs for 3<br />

months. The lipid profiles of both the groups were compared at the start and at the end of 3<br />

months.<br />

Results: After intervention with yoga for a period of 3 months the study group showed a<br />

decrease in total cholesterol, triglycerides and LDL, with an improvement in HDL.<br />

Conclusion: Yoga, being a lifestyle incorporating exercise and stress management training,<br />

targets the elevated lipid levels in patients with diabetes through integrated approaches.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

1. Introduction<br />

The major risk factor for cardiovascular disease in diabetes<br />

mellitus is dyslipidemia. 1 The characteristic features of diabetic<br />

dyslipidemia are a high plasma triglyceride concentration,<br />

low high-density lipoprotein (HDL) concentration and<br />

increased concentration of small dense low-density lipoprotein<br />

(LDL) particles. Insulin resistance leads to increased flux<br />

of free fatty acids and hence the lipid changes. 2 Reports from<br />

the National Health and Nutrition Examination Survey<br />

(NHANES) 1999e2000 indicate that 55% of the U.S. general<br />

population and 51% of adults aged 20e59 years with diabetes<br />

have hypercholesterolemia. 3,4<br />

Caloric restriction and weight loss for the overweight<br />

individual with diabetes mellitus have been of proven<br />

therapeutic value. However, there is no consensus on the ideal<br />

dietary composition for these patients. Genetic factors and the<br />

lipid phenotype of the individual determine the way the<br />

plasma lipid profiles change in these patients. 5 Lifestyle<br />

changes, including increased physical activity and dietary<br />

modifications have, however, been the cornerstones of management<br />

of dyslipidemia in diabetes. 6<br />

Mahajan conducted a study on subjects with mild to<br />

moderate hypertension and reported that yoga can play an<br />

important role in risk modification for cardiovascular diseases.<br />

7 Another study had reported a better lipid profile in<br />

long and medium term mediators when compared to nonmeditators.<br />

8 In view of these observations, the present study<br />

was undertaken to assess the effect of yoga practice on the<br />

lipid profile in patients with type 2 diabetes mellitus (DM).<br />

* Corresponding author.<br />

E-mail address: nisha@gmu.ac.ae (N. Shantakumari).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.010


128<br />

indian heart journal 65 (<strong>2013</strong>) 127e131<br />

2. Materials and methods<br />

2.1. Setting<br />

Patients reporting to the out-patient department of Holistic<br />

Medicine and in the Diabetic clinic, Medical College Trivandrum,<br />

Kerala, India, in the year 2005 were the participants of<br />

this randomized parallel study. The Holistic Medicine<br />

department aims at bringing lifestyle modifications in patients<br />

with non-communicable diseases and also offers regular<br />

yoga classes for all the patients in its care. The study was<br />

conducted after the approval of the Ethics Committee of the<br />

Medical faculty and all subjects volunteered for the trial.<br />

2.2. Study sample<br />

The guidelines of the National Diabetes Data Group and the<br />

third set of the Adult Treatment Panel of the National<br />

Cholesterol Education Program (NCEP ATP III) were used to<br />

recruit one hundred patients with type 2 diabetes and dyslipidemia.<br />

9,10 Known diabetic patients who were on treatment<br />

with sulphonylureas were included in the study. Patients who<br />

are smokers, alcoholics, pregnant, on long-term steroids and<br />

those with known retinopathy, nephropathy, coronary artery<br />

disease and cerebrovascular diseases were excluded from the<br />

study.<br />

One hundred patients with type 2 DM attending the Diabetic<br />

clinic were randomized into control group of 27 males<br />

and 23 females and experimental group of 24 males and 26<br />

females. The experimental group was prescribed oral hypoglycemic<br />

drugs and in addition followed lifestyle modification<br />

in the form of 1 h daily practice of yoga for a period of 3<br />

months at the Holistic Medicine department. The control<br />

group was prescribed oral hypoglycemic drugs only and did<br />

not perform yogic exercises during this period.<br />

The experimental group was taught a series of yoga postures<br />

in groups of 25 patients each. They were instructed to practise<br />

them daily for 1 h duration and were asked to record the<br />

number of minutes they engaged in yoga per day. Yoga<br />

treatment consisted of practice of 1) asanas (Body postures), 2)<br />

pranayama (Breathing exercises) and 3) meditation techniques<br />

(Table 1), to be practiced for 1 h duration. At the end of<br />

2 weeks of supervised yogic training each subject was given<br />

advice on ongoing medical treatment and was given a booklet<br />

illustrating personalized yoga program to practice regularly at<br />

home. All subjects were required to contact the Holistic<br />

Medicine department once every month for follow up advice.<br />

The control group was asked to report to Diabetic clinic every<br />

month for their follow up after being given advice on ongoing<br />

medical treatment at the start of the study. There were no<br />

alterations made in the treatment and dietary habits of either<br />

group during the study period. Both the groups were advised<br />

to continue with their carbohydrate restricted fiber rich diet.<br />

The BMI, W/H circumference and lipid profile of all the<br />

participants were measured at the end of 3 months. Data were<br />

entered in Microsoft Excel and SPSS Version 12 was used for<br />

analysis. Paired and unpaired t tests were employed to<br />

compare measures. A p value of


indian heart journal 65 (<strong>2013</strong>) 127e131 129<br />

Table 2 e Baseline characteristics of the participants.<br />

Parameters Control group Study group<br />

The study showed that 3 months of yoga practice resulted<br />

in a non-significant decrease in BMI from 25.12 1.54 to<br />

23.59 1.38 kg/m 2 . A significant decrease in the weight from<br />

62.20 4.45 to 59.60 4.65 kg and W/H ratio from 0.94 þ 0.07 to<br />

0.89 þ 0.07 was recorded.<br />

There was a significant reduction in total cholesterol, triglycerides<br />

and LDL cholesterol. Mean total cholesterol before<br />

yoga was 244.86 28.09 mg% and was reduced to a mean of<br />

219.56 32.02 mg%. Triglycerides showed a significant<br />

reduction from 151.88 43.08 mg% to 130.11 28.82 mg%<br />

while the LDL reduced from 144.74 28.45 to 120.51 34.31 mg<br />

%. There was a non-significant elevation in HDL from<br />

44.63 9.35 mg% to 47.15 8.17 mg % (Table 3).<br />

After a period of 3 months the control group showed a<br />

significant increase in body weight, non-significant increase<br />

in BMI, total cholesterol, triglycerides and LDL and a decrease<br />

in HDL (Table 4).<br />

4. Discussion<br />

(Medication<br />

only) n ¼ 50<br />

(Medication þ<br />

Yogic intervention)<br />

n ¼ 50<br />

Mean age (years) 44.46 10.98 45.51 7.98<br />

Sex (M/F) 23 F, 27 M 26 F, 24 M<br />

Wt (kg) 62.20 4.45 62.17 4.67<br />

BMI (kg/m 2 ) 23.2 2.14 22.9 2.15<br />

W/H ratio 0.93 þ 0.07 0.94 þ 0.07<br />

FBS (mg/dl) 181.57 66.25 155.86 60.53<br />

PPBS (mg/dl) 265.31 74.70 240.31 79.42<br />

Total cholesterol (mg/dl) 225.74 37.60 244.86 28.09*<br />

Triglyceride (mg/dl) 172.74 52.55 151.89 43.08<br />

LDL cholesterol (mg/dl) 126.11 30.41 144.74 28.46*<br />

HDL cholesterol (mg/dl) 44.23 5.21 44.63 9.35<br />

*p < 0.05.<br />

The present study was aimed at studying the effect of practicing<br />

yoga in patients with type 2 DM for 3 months. The<br />

practice of yoga in these patients resulted in a decrease in BMI,<br />

body weight, W/H ratio, total cholesterol, triglycerides and<br />

LDL cholesterol and an increase in HDL.<br />

Table 3 e Comparison of pre-yoga and post-yoga values<br />

in experimental group.<br />

Parameters n ¼ 50<br />

Pre-yoga<br />

(mean SD)<br />

Post-yoga<br />

(mean SD)<br />

WT(kg) 62.20 4.45 59.60 4.65*<br />

BMI (kg/m 2 ) 25.12 1.54 23.59 1.38<br />

W/H ratio 0.94 þ 0.07 0.89 þ 0.07*<br />

Total cholesterol (mg/dl) 244.86 28.09 219.54 32.02**<br />

Triglycerides (mg/dl) 151.88 43.08 130.11 28.82*<br />

LDL cholesterol (mg/dl) 144.74 28.45 120.51 34.31**<br />

HDL cholesterol (mg/dl) 44.63 9.35 47.15 8.17<br />

*p < 0.05, **p < 0.01.<br />

Table 4 e Comparison of initial values of parameters and<br />

follow up values of the control group.<br />

Parameters n ¼ 50<br />

Initial value<br />

(mean SD)<br />

Follow up<br />

(mean SD)<br />

WT (kg) 62.17 4.67 63.03 5.10*<br />

BMI (kg/m 2 ) 24.73 1.87 25.03 2.14<br />

W/H ratio 0.93 þ 0.07 0.91 þ 0.05<br />

Total cholesterol (mg/dl) 225.74 37.60 235.23 26.64<br />

Triglycerides (mg/dl) 172.74 52.55 197.91 130.11<br />

LDL cholesterol (mg/dl) 126.11 30.41 126.60 22.84<br />

HDL cholesterol (mg/dl) 44.23 5.21 43.13 12.33<br />

*p < 0.05.<br />

Improving glycemic control has not yet been shown to<br />

prevent development of macro-vascular complications in type<br />

2 DM. Alternately, carefully controlled treatment measures<br />

with exercise, dietary modification and oral drugs can be expected<br />

to improve diabetic lipid disorder. 14<br />

The effect of exercise on blood lipid profiles has been<br />

widely reported. 15,16 Physical activity raises HDL levels and<br />

decreases the concentration of very low-density lipoprotein<br />

cholesterol and triglycerides. 17 Physical activity and HDL<br />

appear to be linked via HDL’s role in triglyceride metabolism. 18<br />

It is, however, seen that diabetic patients usually cannot<br />

sustain the levels of recommended physical activity for them<br />

due to varied reasons like age, obesity, cardiovascular disease<br />

and other complications. Compliance and motivation for<br />

performing activity at 50e70% of maximum aerobic capacity<br />

regularly is quite poor. 19<br />

Yoga has a beneficial effect on insulin kinetics and the lipid<br />

profile resulting from it. A decrease inwaist hip ratio inthis study<br />

is consistent with that reported by Sahay et al, who also reported<br />

an associated increase in lean body mass and reduction in skin<br />

fold thickness. Yoga helps in redistribution of body fat and<br />

reduction in central obesity which leads to insulin resistance. A<br />

decrease in insulin resistance, increase in insulin receptors and<br />

sensitivity,shift of peak level of insulinto leftwithnormalization<br />

of insulin glucagon ratio was also reported. 20<br />

The dynamic stretching of the body during yoga asanas is<br />

postulated to rejuvenate pancreatic cells, increase insulin<br />

secretion and hence correct the impaired insulin secretion in<br />

chronic diabetes. 21<br />

Various studies have reported physical, physiological,<br />

psychological and endocrinological changes with the practice<br />

of yoga. Manchanda and Narang found that after one year of<br />

yoga therapy, patients with coronary artery disease (CAD)<br />

showed significant reduction in serum total cholesterol, triglycerides<br />

and LDL cholesterol. The patients showed a significant<br />

reduction in the number of angina episodes and<br />

required revascularization procedures less frequently. Angiography<br />

after one year showed regression of lesions. 22 The<br />

practice of Raja yoga meditation was found to lower serum<br />

cholesterol by Vyas, 23 while Sahay 20 and Bijlani 24 noted that<br />

yoga practice causes significant reduction in free fatty acids,<br />

LDL, VLDL and an increase in HDL.<br />

The beneficial effect of yoga in the management of<br />

hyperlipidemia and obesity cannot just be attributed alone to<br />

the simple excess calorie expenditure as there is no rapid<br />

muscle activity and energy generation involved in yoga.


130<br />

indian heart journal 65 (<strong>2013</strong>) 127e131<br />

Repeated stress is known to lead to persistent elevation of<br />

cortisol which causes central obesity and insulin resistance. 25<br />

It increases gluconeogenesis and diminishes peripheral uptake<br />

of glucose. 26 Decreased glucose uptake and insulin<br />

secretion can also occur due to stress induced release of<br />

growth hormone and b endorphin. 27 Elevated cortisol is also<br />

linked to dyslipidemia, and higher blood pressure. 28 Yoga has<br />

been reported to lower levels of sympathetic hormones and<br />

reduce cortisol. Pranayama reduces sympathetic tone, increases<br />

parasympathetic activity and also helps an individual<br />

reduce stress. 8,29,30 Meditation also brings about a hypometabolic<br />

state and reduces stress induced sympathetic over<br />

activity. 31 Better ability to overcome stress resulting in lowered<br />

cortisol levels can be cited as possible mechanism for<br />

improvement in lipid profile in patients practicing yoga.<br />

Dyslipidemia is usually associated with the abnormalities<br />

in lipolysis; triglyceride metabolism and free fatty acid turn<br />

over in a case of insulin resistance. Impaired lipoprotein lipase<br />

and increased hepatic lipase activity is thought be a resulting<br />

from insulin resistance in diabetes. Chronic exposure to<br />

elevated free fatty acids have been associated with impaired<br />

insulin secretion. 32 The improvement in lipid profile with<br />

practice of yoga could be due to increased hepatic lipase and<br />

lipoprotein lipase. This would increase the uptake of triglycerides<br />

by adipose tissue and affect the lipoprotein<br />

metabolism. 33<br />

Yoga practice is also proved to affect mental balance of an<br />

individual allaying apprehension, stress and bringing about<br />

hormonal balance and feelings of well being. This sense of<br />

well being is attributed to its ability to increase endogenous<br />

melatonin secretion. 34 This can explain the probability of<br />

greater compliance with its practice even in long-term and its<br />

use as an effective intervention in control of the disease.<br />

5. Conclusion<br />

The present study has shown an efficacy of improving the<br />

dyslipidemic state associated with diabetes. Yoga, being a<br />

lifestyle incorporating exercise and stress management<br />

training, targets the elevated lipid through integrated approaches<br />

resulting in improved lipid profiles, lower BMI, and<br />

macro-vascular complications in diabetes.<br />

5.1. Strength of study<br />

The yoga package was designed after extensive literature review<br />

by yoga specialists and was a perfect combination of<br />

asana and breathing exercises targeted at the disease under<br />

study. Excellent compliance of study sample and there were<br />

no drop outs. Experimental group patients voluntarily reported<br />

to Holistic medicine department and were self motivated<br />

for the practice of yoga. The control group was also<br />

under constant surveillance by the Diabetic care clinic.<br />

5.2. Limitations of the study<br />

Direct supervision of the patients was not possible for the<br />

entire period of the study. Dietary data were not recorded.<br />

Long-term study was not possible due to threat of noncompliance<br />

of the patients.<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Taskinen MR. Diabetic dyslipidemia. Atheroscler Suppl.<br />

2002;3(1):47e51.<br />

2. Mooradian Arshag D. Dyslipidemia in type 2 diabetes<br />

mellitus. Nat Clin Pract Endocrinol Metab. 2009;5:150e159.<br />

3. Ford ES, Mokdad AH, Giles WH, Mensah GA. Serum total<br />

cholesterol concentrations and awareness, treatment, and<br />

control of hypercholesterolemia among US adults: findings<br />

from the National Health and Nutrition Examination Survey,<br />

1999 to 2000. Circulation. 2003;107:2185e2189.<br />

4. Imperatore G, Cadwell BL, Geiss L, et al. Thirty year trends in<br />

cardiovascular risk factor levels among US adults with<br />

diabetes: National Health and Nutrition Examination Surveys,<br />

1971e2000. Am J Epidemiol. 2004;160:531e539.<br />

5. Krauss RM. Dietary and genetic probes of atherogenic<br />

dyslipidemia. Arterioscler Thromb Vasc Biol. 2005;25:<br />

2265e2272.<br />

6. Expert Panel on Detection, Evaluation, and Treatment of High<br />

Blood Cholesterol in Adults. Executive summary of the third<br />

report of the National cholesterol education program (NCEP)<br />

expert Panel on detection, evaluation, and treatment of high<br />

blood cholesterol in adults (Adult treatment Panel III). JAMA.<br />

2001;285:2486e2497.<br />

7. Mahajan AS, Reddy KS, Sachdeva U. Lipid profile of coronary<br />

risk subjects following yogic lifestyle intervention. <strong>Indian</strong><br />

<strong>Heart</strong> J. 1999;51:37e40.<br />

8. Damodaran A, Malathi A, Patil N, Shah N, Suryananshi,<br />

<strong>Mar</strong>athe S. Therapeutic potential of yoga practices in<br />

modifying cardiovascular risk profile in middle aged men and<br />

women. J Assoc Physicians India. 2002;50:633e640.<br />

9. Vyas R, Dikshit N. Effect of meditation on respiratory system,<br />

cardiovascular system and lipid profile. Ind J Physiol Pharmacol.<br />

2002;46(4):487e491.<br />

10. National Diabetes Data Group. Classification and diagnosis of<br />

diabetes mellitus and other categories of glucose intolerance.<br />

Diabetes. 1979;28:1039e1057.<br />

11. Expert Panel on Detection, Evaluation and treatment of high<br />

blood cholesterol in adults (Adult treatment Panel III). JAMA.<br />

2001;285:2486e2497.<br />

12. Mc Gowan MW, Artiss JD, Strandbergh DR, Zak B. A peroxidasecoupled<br />

method for the colorimetric determination of serum<br />

triglycerides. Clin Chem. 1983;29:538e542.<br />

13. Trider P. Clinical chemistry. Ann Clin Biochem. 1969;6:24e27.<br />

14. Burstein M, Scholnic HR, Morfin R. Rapid method for the<br />

isolation of lipoproteins from human serum with polyanions.<br />

J Lipid Res. 1970;11:583e595.<br />

15. Pyorala k, Laakso M, Ustipa M. Diabetes and atherosclerosis;<br />

an epidemiological view. Diabetes Metab Rev. 1987;3:463e524.<br />

16. Szapary PO, Bloedon LT, Foster GD. Physical activity and its<br />

effects on lipids. Curr Cardiol Rep. 2003;5:488e492.<br />

17. Asikainen TM, Miilunpalo S, Kukkonen-Harjula K, et al.<br />

Walking trials in postmenopausal women: effect of low doses<br />

of exercise and exercise fractionization on coronary risk<br />

factors. Scand J Med Sci Sports. 2003;13:284e292.<br />

18. Clearfield Michael B. The national cholesterol education<br />

program adult treatment Panel III guidelines. JAOA; 2003.


indian heart journal 65 (<strong>2013</strong>) 127e131 131<br />

19. Thompson PD. What do muscles have to do with<br />

lipoproteins? Circulation. 1990;81:1428e1430.<br />

20. Skarfors ET, Wegener TA, Lithell H, Selinus I. Physical training<br />

as treatment for Type 2 (non-insulin-dependent) diabetes in<br />

elderly men. A Feasibility Study Over 2 Years. Diabetologia.<br />

1987;30(12):930e933.<br />

21. Sahay BK. Role of yoga in diabetes. J Assoc Physicians India.<br />

2007;55:121e126.<br />

22. Singh S, Malhotra V, Singh KP, Madhu SV, Tandon OP. Role of<br />

yoga in modifying certain cardiovascular functions in Type 2<br />

diabetic patients. J Assoc Physicians India. 2004;52:203.<br />

23. Manchnada SC, Narang R, Reddy KS, Sachdev V. Retardation<br />

of coronary atherosclerosi with yoga lifestyle intervention.<br />

J Asso Physiocians in India. 2000;48:487e494.<br />

24. Vyas R, Dikshit N. Effect of meditation on respiratory system,<br />

cardiovascular system and lipid profile. <strong>Indian</strong> J Physio<br />

Pharmacol. 2001;45(4):493e496.<br />

25. Bijlani RL, Vempati RP, Yadav RK, et al. A brief but<br />

comprehensive lifestyle education program based on yoga<br />

reduces risk factors for cardiovascular disease and diabetes<br />

mellitus. J Altern Complement Med. 2005;11(2):267e274.<br />

26. Bjorntorp P. Metabolic implications of body fat distribution.<br />

Diabetes Care. 1991;14:1132e1143.<br />

27. Chrousos GP, Gold PW. A healthy body in a healthy mind and<br />

vice versa: the damaging power of uncontrollable stress. J Clin<br />

Endocrinol Metab. 1998;83:1842e1845.<br />

28. Giugliano D. Morphine, opioid peptides, and pancreatic islet<br />

function. Diabetes Care. 1984;7:92e98.<br />

29. Udupa K, Madanmohan, Ananda BB, Vijayalakshmi P,<br />

Krishnamoorthy N. Effect of pranayama training on cardiac<br />

function in normal young volunteers. <strong>Indian</strong> J Physiol<br />

Pharmacol. 2003;47:27e33.<br />

30. Raghuraj P, Ramakrishnan AG, Nagendra HR, Telles S. Effect<br />

of two selected yogic breathing techniques on heart rate<br />

variability. <strong>Indian</strong> J Physiol Pharmacol. 1998;42:467e472.<br />

31. Sandeep B, Pandey US, Verma NS. Improvement in oxidative<br />

status with yogic breathing in young healthy males. <strong>Indian</strong> J<br />

Physiol Pharmacol. 2002;46:349e354.<br />

32. Perez-De-Albeniz A, Holmes J. Meditation:concepts, effects<br />

and uses in therapy. Int J Psychotherapy. 2000;5:49e58.<br />

33. Shradha Bisht, Sisodia SS. Diabetes, dyslipidemia, antioxidant<br />

and status of oxidative stress. IJRAP. 2010;1(1):33e42.<br />

34. Harinath K, Malhotra AS, Pal K, et al. Effects of hatha yoga<br />

and omkar meditation on cardiorespiratory performance,<br />

psychological profile and melatonin secretion. J Altern<br />

Complement Med. 2004;10(2):261e268.


indian heart journal 65 (<strong>2013</strong>) 137e141<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Comparison of diagnostic utilities of ankleebrachial index<br />

and Carotid intima-media thickness as surrogate markers of<br />

significant coronary atherosclerosis in <strong>Indian</strong>s<br />

Babu Ezhumalai c, *, Subrahmanyam Dharanipragada Krishnasuri b ,<br />

Balachander Jayaraman a<br />

a Dept. of Cardiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605006, India<br />

b Dept. of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605006, India<br />

c Senior Resident, Dept. of Cardiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantri Nagar, Gorimedu,<br />

Pondicherry 605006, India<br />

article info<br />

Article history:<br />

Received 4 June 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 21 February <strong>2013</strong><br />

Keywords:<br />

Coronary artery disease<br />

Ankleebrachial index<br />

Carotid intima-media thickness<br />

Atherosclerosis<br />

abstract<br />

Aim: We aimed to compare Ankleebrachial index (ABI) and Carotid intima-media thickness<br />

(CIMT) as surrogate markers of significant coronary atherosclerosis in South <strong>Indian</strong>s with<br />

coronary artery disease (CAD).<br />

Methods and results: There were two groups: CAD group (n ¼ 59) and Control group (n ¼ 55).<br />

Mean ABI (0.82 0.06 vs. 1.16 0.11, p < 0.0001) and mean CIMT (0.74 0.22 mm vs.<br />

0.45 0.09 mm, p < 0.0001) were statistically different between two groups. ABI < 0.9<br />

(sensitivity: 91.53%, specificity: 100%) and CIMT > 0.63 mm (sensitivity: 61.02%, specificity:<br />

98.18%) implied significant CAD. ABI and CIMT were negatively correlated to one another.<br />

With increasing severity of CAD, ABI decreased but CIMT increased.<br />

Conclusion: ABI and CIMT are simple noninvasive tools providing insight into coronary<br />

atherosclerosis. They can be done at bedside and easily repeated than coronary angiography.<br />

ABI < 0.9 is a better surrogate marker of significant coronary atherosclerosis than<br />

CIMT > 0.63 mm in South <strong>Indian</strong>s with CAD.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

1. Introduction<br />

Coronary artery disease (CAD) imposes significant economic<br />

burden throughout the world. Coronary angiography remains<br />

the gold standard investigation for identifying CAD. In fact, it<br />

is a luminogram and may not detect the abluminal plaque<br />

growth in the initial stages of coronary atherosclerosis.<br />

Moreover, it is invasive associated with significant interobserver<br />

variability and underestimation of stenosis in the<br />

presence of diffuse disease.<br />

Atherosclerosis is the major etiology of CAD and it is a<br />

generalized process simultaneously involving multiple arteries<br />

in the body. Hence, analysis of peripheral arteries in an<br />

individual with CAD may throw some light upon the degree<br />

of atherosclerosis at the level of coronaries. Ankleebrachial<br />

index (ABI) and Carotid intima-media thickness (CIMT) are<br />

simple noninvasive tools developed based on this principle.<br />

The use of ABI in peripheral arterial disease is well established.<br />

As the same pathogenesis primarily involves coronary<br />

vasculature, ABI (0.9 or


138<br />

indian heart journal 65 (<strong>2013</strong>) 137e141<br />

cardiovascular events and death. 1e3 Abnormal ABIs, both low<br />

(


indian heart journal 65 (<strong>2013</strong>) 137e141 139<br />

The differences in mean ABI (CAD group: 0.82 0.06 and<br />

Control group: 1.16 0.11, unpaired Student’s t test p < 0.0001)<br />

and mean CIMT (CAD group: 0.74 0.22 mm and Control<br />

group: 0.45 0.09 mm, unpaired Student’s t test p < 0.0001)<br />

between the two groups were statistically highly significant<br />

(Table 2). Overall, in the CAD group, there were 54 (91.53%)<br />

cases with ABI < 0.9 and 5 (8.47%) cases with ABI 0.9 (Table<br />

2). Similarly, there were 36 (61.02%) cases with<br />

CIMT > 0.63 mm and 23 (38.98%) cases with CIMT 0.63 mm in<br />

the CAD group. There were 22 (37.29%) cases with carotid<br />

plaques in CAD group while none in the Control group had<br />

plaque. There was significant difference (Fisher’s Exact test,<br />

p < 0.0001) between the two groups with respect to the presence<br />

of carotid plaques.<br />

Subgroup analysis (Table 3) between diabetics (n ¼ 28) and<br />

non-diabetics (n ¼ 31) within CAD group did not show significant<br />

difference in the mean values of ABI (0.82 0.06 and<br />

0.82 0.05, p ¼ 0.714) or CIMT (0.75 0.24 and 0.73 0.20,<br />

p ¼ 0.679) using unpaired Student’s t test.<br />

The mean ABI was 0.87, 0.82 and 0.75 in cases with single<br />

vessel, double vessel and triple vessel diseases in CAD group<br />

respectively. One-way ANOVA showed that these values were<br />

statistically different (F ¼ 42.487, p < 0.0001) between these<br />

subgroups. The mean CIMT was 0.6, 0.73 and 0.94 mm in these<br />

subgroups respectively and they were statistically different<br />

(F ¼ 44.552, p < 0.0001, one-way ANOVA). The posthoc test<br />

performed along with ANOVA was Turkey Kramer Multiple<br />

Comparisons test and this also showed statistical significance<br />

( p < 0.001) with respect to ABI and CIMT in these subgroups.<br />

With increase in severity of CAD, ABI decreased (Fig. 1) while<br />

CIMT increased (Fig. 2). Pearson correlation test showed statistically<br />

significant negative correlation between ABI and<br />

CIMT (r ¼ -0.780).<br />

5. Discussion<br />

Routine screening tests such as resting electrocardiography,<br />

treadmill test and electron-beam computed tomography are<br />

inaccurate in predicting cardiovascular events in adults with<br />

low risk for CAD or asymptomatic persons because many<br />

acute events result from sudden occlusion of a previously<br />

unobstructed coronary artery. 12 Therefore, in our study, the<br />

participants in Control group were not routinely subjected to<br />

Table 2 e Ankleebrachial index and Carotid intimamedia<br />

thickness in CAD and Control groups.<br />

Parameter<br />

CAD group<br />

n ¼ 59<br />

Control group<br />

n ¼ 55<br />

ABI<br />

Mean SD* 0.82 0.06 1.16 0.11<br />

0.63 36 (61.02%) 1 (1.82%)<br />

0.63 23 (38.98%) 54 (98.18%)<br />

*p < 0.0001 using unpaired Student’s t test.<br />

Table 3 e Ankleebrachial index and carotid intima-media<br />

thickness between diabetics and non-diabetics within<br />

CAD group.<br />

Parameter<br />

Diabetics<br />

n ¼ 28<br />

Mean SD<br />

Non-diabetics<br />

n ¼ 31<br />

Mean SD<br />

ABI* 0.82 0.06 0.82 0.05<br />

CIMT # (mm) 0.75 0.24 0.73 0.20<br />

*p ¼ 0.714 and # p ¼ 0.679 using unpaired Student’s t test.<br />

these screening tests but recruited based on Framingham risk<br />

scoring with 10-year risk of developing myocardial infarction<br />

and coronary death less than 10%.<br />

Prior studies have concluded that a low ABI is highly specific<br />

but not sensitive for predicting future cardiovascular risk<br />

in individuals with peripheral arterial diseases. 13 There is one<br />

trial where ABI was assessed using automated oscillometric<br />

method and it was found that the sensitivity, specificity, and<br />

positive and negative predictive values of ABI < 0.9 in predicting<br />

multivessel CAD were 22%, 96%, 93%, and 34%,<br />

respectively. 14 In contrast with this trial, another study done<br />

in African-American population, proved that an ABI 0.90<br />

predicted the presence of 3-vessel or left main CAD with a<br />

sensitivity of 85% and specificity of 77%. 15 In patients with<br />

PAD diagnosed based on ABI < 0.9, the prevalence of CAD is<br />

46.88%. 16<br />

The normal range (mean 2SE, 95% confidence interval) of<br />

ABI for healthy adults in our study is from 0.94 to 1.38. Taking<br />

0.94 as the lower limit of normal range for ABI, we would obtain<br />

a sensitivity of 100% and specificity of 96.36%. Since many<br />

studies have shown that ABI < 0.9 is a marker of increased<br />

cardiovascular events, 1e3 we took ABI < 0.9 as a surrogate<br />

marker of CAD and found a sensitivity of 91.53%, specificity of<br />

100%, pretest probability of 51.75%, positive predictive value of<br />

100% and a negative predictive value of 91.67%.<br />

0.9<br />

0.85<br />

0.8<br />

Mean<br />

ABI<br />

5<br />

0.7<br />

0.65<br />

0.87<br />

Single Vessel<br />

Disease<br />

0.82<br />

Double Vessel<br />

Disease<br />

0.75<br />

Triple Vessel<br />

Disease<br />

Fig. 1 e Ankleebrachial index and severity of coronary<br />

artery disease. One-way ANOVA: F [ 42.48, p < 0.0001.<br />

Posthoc test: TurkeyeKramer Multiple Comparisons test,<br />

p < 0.001.


140<br />

indian heart journal 65 (<strong>2013</strong>) 137e141<br />

1<br />

0.9<br />

0.8<br />

Mean<br />

CIMT 0.7<br />

(mm) 0.6<br />

0.6<br />

0.5<br />

0.4<br />

Single Vessel<br />

Disease<br />

0.73<br />

Double Vessel<br />

Disease<br />

0.94<br />

Triple Vessel<br />

Disease<br />

Fig. 2 e Carotid intima-media thickness and severity of<br />

coronary artery disease. One-way ANOVA: F [ 44.552,<br />

p < 0.0001. Posthoc test: TurkeyeKramer Multiple<br />

Comparisons test, p < 0.001.<br />

Prior studies had no unanimous opinion about the cut-off<br />

value of CIMT for CAD. According to one study conducted in<br />

Japanese diabetics, CIMT 1.1 mm represents CAD. 10 The<br />

maximum values of CIMT for normal <strong>Indian</strong> population and<br />

CAD are 0.80 and 1.02 mm, respectively. 17 There is one more<br />

study which states that the risk of first myocardial infarction<br />

increases with a CIMT 0.822 mm. 18 In our study, the<br />

normal range (mean 2SE, 95% confidence interval) of CIMT<br />

for healthy adults is from 0.27 to 0.63 mm. We took<br />

CIMT > 0.63 mm as a surrogate marker of CAD and found a<br />

sensitivity of 61.02%, specificity of 98.18%, pretest probability<br />

of 51.75%, positive predictive value of 97.3% and a negative<br />

predictive value of 70.13%. In published studies, CIMT cut-off<br />

value of 0.67 mm yielded a sensitivity and specificity of 85%<br />

and 72%, respectively, for predicting obstructive coronary<br />

atherosclerosis 19 while the cut-off value of 0.87 mm for<br />

maximum CIMT obtained a sensitivity and specificity of 90<br />

and 64%, respectively for the detection of left main CAD. 5 In<br />

our study, when a higher cut-off value of 0.8 mm was<br />

considered the upper limit of normal CIMT, specificity<br />

increased to 100% but sensitivity decreased markedly to<br />

16.95%. Hence we took 0.63 mm as the upper limit of normal<br />

CIMT. This cut-off value is lower than that found in other<br />

studies. The probable explanation for a lower cut-off value for<br />

CIMT in our study is that other studies had matched cases and<br />

controls with respect to risk factors like diabetes, hypertension,<br />

dyslipidemia, smoking, obesity etc. On the other hand,<br />

the controls in our study were matched with cases only in age<br />

and sex but not for risk factors. Patients with diabetes were<br />

excluded from the control population. Moreover, the inclusion<br />

criteria for Control group incorporated Framingham risk<br />

scoring to calculate 10-year risk of developing cardiovascular<br />

events less than 10% and this was hardly used by other<br />

studies. Another explanation could be that South <strong>Indian</strong>s may<br />

have a lower CIMT than other populations. Our study is<br />

limited by small sample size which could have also contributed<br />

to the lower cut-off value for CIMT.<br />

Based on sensitivity and specificity, ABI < 0.9 is a better<br />

surrogate marker of CAD than CIMT > 0.63 mm. In our study,<br />

all assessments were performed by a single operator so as to<br />

avoid interobserver variation and multiple readings were<br />

taken in order to avoid intraobserver variation. Earlier done<br />

study also did not show any significant interobserver or<br />

intraobserver variation on repeatedly measuring ABI. 20<br />

Studies have shown that significant reduction in CIMT occurs<br />

by one year of therapy with statins 21 and maximum<br />

regression occurs by 2 years. 22 So we excluded all those cases<br />

on statins for more than one year.<br />

As per ATP III guidelines, diabetes mellitus is considered a<br />

CAD equivalent. 11 Hence, ABI is expected to be low and CIMT to<br />

be high in diabetics compared to non-diabetics. The abnormalities<br />

in ABI and CIMT are expected to be more pronounced<br />

in diabetics with CAD than in non-diabetics with CAD. On the<br />

contrary, in our study, there was no significant difference in<br />

ABI or CIMT between diabetics and non-diabetics within CAD<br />

group. The major reason for this interesting observation is the<br />

quantum of atherosclerosis needed for developing CAD will<br />

not differ between diabetics and non-diabetics. Other reasons<br />

could be the interplay of multiple risk factors in non-diabetics<br />

within CAD group and small sample size.<br />

As mentioned earlier, in our study, the term ‘severity’ of<br />

CAD referred only to the number of major epicardial coronary<br />

arteries having at least 50% stenosis. We did not take into<br />

consideration the degree of stenosis beyond 50% and the type<br />

of coronary artery involved. There was significant difference<br />

in ABI and CIMT among cases with single vessel disease,<br />

double vessel disease and triple vessel disease within the CAD<br />

group. With increase in severity of CAD, ABI decreased but<br />

CIMT increased in our study. Prior studies had shown similar<br />

variation of ABI and CIMT with respect to severity of<br />

CAD. 8,10,23e25 A few studies differed in this view; one study<br />

found a weak correlation between CIMT and severity of CAD 26<br />

and a couple of studies could not establish a direct association<br />

between ABI and significant CAD. 27,28 We also found that ABI<br />

and CIMT were negatively correlated to one another; i.e. with<br />

increase in CIMT there was a decrease in ABI and vice versa.<br />

The definition of carotid plaque is highly variable. In our<br />

study, carotid plaque was defined as a localized thickening of<br />

the intima of carotid artery greater than 1.5 mm. 5,29 Carotid<br />

arteries were scanned from the clavicle to the angle of<br />

mandible for luminal plaques, while maximal CIMT was<br />

measured in the far wall of common carotid artery around<br />

10 mm below the bifurcation. Around 37.29% of cases in CAD<br />

group had plaques in carotid artery while none in the Control<br />

group had plaques. Using Fisher’s exact test we found that<br />

there was highly significant difference between CAD and<br />

Control groups with respect to the presence of carotid plaques.<br />

Hence, the value of CIMT as a surrogate marker of CAD is<br />

emphasized by the presence of plaque in carotid arteries.<br />

6. Conclusion<br />

Ankleebrachial index and Carotid intima-media thickness are<br />

simple noninvasive diagnostic tools capable of providing<br />

insight into the burden of atherosclerosis at the level of coronaries.<br />

Hence they are surrogate markers of significant


indian heart journal 65 (<strong>2013</strong>) 137e141 141<br />

coronary atherosclerosis. They are very simple to do at<br />

bedside and more easily repeatable than coronary angiography.<br />

They provide key information even during the early<br />

phase of coronary atheroma formation particularly during the<br />

stage of positive remodeling when the atheromatous plaque<br />

abluminally encroaches on the coronary arteries. This<br />

compensatory hyperenlargement is usually not discernible in<br />

coronary angiography. Moreover there is no place for issues<br />

like radiation hazard, contrast induced nephropathy or<br />

vascular complications which may be encountered during<br />

coronary angiography. ABI < 0.9 is a better surrogate marker<br />

of significant coronary atherosclerosis than CIMT > 0.63 mm<br />

in South <strong>Indian</strong>s with CAD. The presence of carotid plaque<br />

may endorse CIMT as a surrogate marker of CAD. ABI and<br />

CIMT are negatively correlated to one another. Therefore, ABI<br />

decreases but CIMT increases with increasing burden of coronary<br />

atherosclerosis.<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Lee AJ, Price JF, Russell MJ, et al. Improved prediction of fatal<br />

myocardial infarction using the ankle brachial index in<br />

addition to conventional risk factors: the Edinburgh Artery<br />

Study. Circulation. 2004;110(19):3075e3080.<br />

2. Leng GC, Fowkes FG, Lee AJ, et al. Use of ankle brachial<br />

pressure index to predict cardiovascular events and death: a<br />

cohort study. BMJ. 1996;313:1440e1444.<br />

3. Papamichael CM, Lekakis JP, Stamatelopoulos KS, et al.<br />

Ankle-brachial index as a predictor of the extent of coronary<br />

atherosclerosis and cardiovascular events in patients with<br />

coronary artery disease. Am J Cardiol. 2000;86(6):615e618.<br />

4. Criqui MH, McClelland RL, McDermott MM, et al. The anklebrachial<br />

index and incident cardiovascular events in the<br />

MESA (Multi-Ethnic Study of Atherosclerosis). J Am Coll<br />

Cardiol. 2010;56(18):1506e1512.<br />

5. Kasliwal RR, Bansal M, Gupta H, et al. Association of carotid<br />

intima-media thickness with left main coronary artery<br />

disease. <strong>Indian</strong> <strong>Heart</strong> J. 2007;59(1):50e55.<br />

6. Wang D, Yang H, Quinones MJ, et al. A genome-wide scan for<br />

carotid artery intima-media thickness: the MexicaneAmerican<br />

Coronary Artery Disease Family Study. Stroke. 2005;36(3):<br />

540e545.<br />

7. Liviakis L, Pogue B, Paramsothy P, et al. Carotid intima-media<br />

thickness for the practicing lipidologist. J Clin Lipidol. 2010;4(1):<br />

24e35.<br />

8. Kablak-Ziembicka A, Tracz W, Przewlocki T, et al. Association<br />

of increased carotid intima-media thickness with the extent<br />

of coronary artery disease. <strong>Heart</strong>. 2004;90(11):1286e1290.<br />

9. Heuten H, Goovaerts I, Ennekens G, et al. Carotid artery<br />

intima-media thickness is associated with coronary artery<br />

disease. Acta Cardiol. 2008;63(3):309e313.<br />

10. Hayashi C, Ogawa O, Kubo S, et al. Ankle brachial pressure<br />

index and carotid intima-media thickness as atherosclerosis<br />

markers in Japanese diabetics. Diabetes Res Clin Pract.<br />

2004;66(3):269e275.<br />

11. Third report of the National Cholesterol Education Program<br />

(NCEP) expert panel on detection, evaluation and treatment<br />

of high blood cholesterol in adults (Adult Treatment Panel III).<br />

Final report. Circulation. 2002;106(25):3143e3421.<br />

12. U.S. Preventive Services Task Force. Screening for coronary<br />

heart disease: recommendation statement. Ann Intern Med.<br />

2004;140(7):569e572.<br />

13. Doobay AV, Anand SS. Sensitivity and specificity of the<br />

ankleebrachial index to predict future cardiovascular<br />

outcomes: a systematic review. Arterioscler Thromb Vasc Biol.<br />

2005;25(7):1463e1469.<br />

14. Su HM, Voon WC, Lin TH, et al. Ankle-brachial pressure index<br />

measured using an automated oscillometric method as a<br />

predictor of the severity of coronary atherosclerosis in<br />

patients with coronary artery disease. Kaohsiung J Med Sci.<br />

2004;20(6):268e272.<br />

15. Otah KE, Madan A, Otah E, et al. Usefulness of an abnormal<br />

ankle-brachial index to predict presence of coronary artery<br />

disease in African-Americans. Am J Cardiol. 2004;93(4):481e483.<br />

16. Sarangi S, Srikant B, Rao DV, et al. Correlation between<br />

peripheral arterial disease and coronary artery disease using<br />

ankle brachial index e a study in <strong>Indian</strong> population. <strong>Indian</strong><br />

<strong>Heart</strong> J. 2012;64(1):2e6.<br />

17. Hansa G, Bhargava K, Bansal M, et al. Carotid intima-media<br />

thickness and coronary artery disease: an <strong>Indian</strong> perspective.<br />

Asian Cardiovasc Thorac Ann. 2003;11(3):217e221.<br />

18. Aminbakhsh A, Mancini GB. Carotid intima-media thickness<br />

measurements: what defines an abnormality? A systematic<br />

review. Clin Invest Med. 1999;22(4):149e157.<br />

19. Djaberi R, Schuijf JD, de Koning EJ, et al. Usefulness of carotid<br />

intima-media thickness in patients with diabetes mellitus as<br />

a predictor of coronary artery disease. Am J Cardiol.<br />

2009;104(8):1041e1046.<br />

20. Endres HG, Hucke C, Holland-Letz T, et al. A new efficient trial<br />

design for assessing reliability of ankle-brachial index<br />

measures by three different observer groups. BMC Cardiovasc<br />

Disord. 2006;6:33.<br />

21. Corti R, Fuster V, Fayad ZA, et al. Effects of aggressive versus<br />

conventional lipid-lowering therapy by simvastatin on<br />

human atherosclerotic lesions: a prospective, randomized,<br />

double-blind trial with high-resolution magnetic resonance<br />

imaging. J Am Coll Cardiol. 2005;46(1):106e112.<br />

22. Crouse JR, Raichlen JS, Riley WA, et al. Effect of rosuvastatin<br />

on progression of carotid intima-media thickness in low-risk<br />

individuals with subclinical atherosclerosis: the METEOR<br />

Trial. JAMA. 2007;297(12):1344e1353.<br />

23. Coskun U, Yildiz A, Esen OB, et al. Relationship between<br />

carotid intima-media thickness and coronary angiographic<br />

findings: a prospective study. Cardiovasc Ultrasound. 2009;7:59.<br />

24. Matsushima Y, Kawano H, Koide Y, et al. Relationship of<br />

carotid intima-media thickness, pulse wave velocity, and<br />

ankle brachial index to the severity of coronary artery<br />

atherosclerosis. Clin Cardiol. 2004;27(11):629e634.<br />

25. Sukhija R, Aronow WS, Yalamanchili K, et al. Association of<br />

ankle-brachial index with severity of angiographic coronary<br />

artery disease in patients with peripheral arterial disease and<br />

coronary artery disease. Cardiology. 2005;103(3):158e160.<br />

26. Adam MR, Nakagomi A, Keech A, et al. Carotid intima-media<br />

thickness is only weakly correlated with the extent and<br />

severity of coronary artery disease. Circulation.<br />

1995;92(8):2127e2134.<br />

27. Hakeem F, Siddique S, Saboor QA. Abnormal ankle brachial<br />

index and the presence of significant coronary artery disease.<br />

J Coll Physicians Surg Pak. 2010;20(2):79e82.<br />

28. Pearson TL. Ankle brachial index as a prognostic tool for<br />

women with coronary artery disease. J Cardiovasc Nurs.<br />

2010;25(1):20e24.<br />

29. Vicenzini E, Ricciardi MC, Puccinelli F, et al. Common carotid<br />

artery intima-media thickness determinants in a population<br />

study. J Ultrasound Med. 2007;26(4):427e432.


indian heart journal 65 (<strong>2013</strong>) 158e167<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Polymorphisms of MDR1, CYP2C19 and P2Y 12 genes in <strong>Indian</strong><br />

population: Effects on clopidogrel response<br />

Kavita K. Shalia d, *, Vinod K. Shah b , Poonam Pawar a , Siddhi S. Divekar a ,<br />

Satchidanand Payannavar c<br />

a Sir H.N. Medical Research Society, Sir H.N. Hospital and Research Centre, Raja Rammohan Roy Road, Mumbai 400 004, India<br />

b Dept. of Cardiology, Sir H.N. Hospital and Research Centre, Raja Rammohan Roy Road, Mumbai 400 004, India<br />

c Dept. of Cardiology, Rajawadi Municipal Hospital, Ghatkopar (East), Mumbai, India<br />

d Research Scientist, Sir H.N. Medical Research Society, Sir H.N. Hospital and Research Centre, Raja Rammohan Roy Road, Mumbai,<br />

Maharashtra 400 004, India<br />

article info<br />

Article history:<br />

Received 29 June 2012<br />

Accepted 14 February <strong>2013</strong><br />

Available online 24 February <strong>2013</strong><br />

Keywords:<br />

Clopidogrel<br />

CYP2C19<br />

Gene polymorphisms<br />

MDR1<br />

P2Y 12<br />

abstract<br />

Aims/objective: Influence of genetic variations on the response of clopidogrel, an antiplatelet<br />

drug is implicated. In the present study, the prevalence of single nucleotide polymorphisms<br />

of MDR1 (C3435T), CYP2C19 [CYP2C19*2 CYP2C19*3, CYP2C19*17] and P2Y 12<br />

(i-T744C) in <strong>Indian</strong> population and their effects on clopidogrel response was analyzed.<br />

Methods and results: To analyze the prevalence of polymorphisms, 102 healthy individuals<br />

were recruited. Clopidogrel response was assessed by ADP induced platelet aggregation in<br />

clopidogrel naïve acute myocardial infarction (AMI) patients (n ¼ 26) screened from 100 AMI<br />

cases, before loading dose of 300 mg, at 24 h before next dose and 6 days after on 75 mg per<br />

day and platelet aggregation inhibition (PAI) was calculated between these time intervals.<br />

Genotyping was carried out by PCR-based restriction enzyme digestion method for C3435T<br />

of MDR1 and i-T744C of P2Y 12 , by multiplex PCR for CYP2C19*2 (G681A) and CYP2C19*3<br />

(G636A) and by nested PCR for CYP2C19*17 (C806T). The effect of the above mentioned<br />

genetic variations on PAI was analyzed. Variant allele of CYP2C19*3 was not observed while<br />

the prevalence of 3435T of MDR1 (0.524), CYP2C19*2 (681A, 0.352); i-744C of P2Y 12 (0.088), as<br />

well as wild type allele CYP2C19*17 (C806, 0.897) associated with decrease clopidogrel<br />

response were observed. Trend toward poor response to clopidogrel was observed at 24 h<br />

with the variant genotypes of CYP2C19*2 and i-T744C of P2Y 12 as compared to wild type.<br />

Conclusion: The present study did show a trend toward impaired response of clopidogrel to<br />

inhibit platelet aggregation with variant genotypes of CYP2C19*2 and iT744C of P2Y 12<br />

compared to respective wild type genotype at 24 h.<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

* Corresponding author. Tel.: þ91 (0) 22 66106308/387; fax: þ91 (0) 22 66106212.<br />

E-mail addresses: Kavita_shalia@hnhospital.com, kavitashalia@hotmail.com (K.K. Shalia).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.012


indian heart journal 65 (<strong>2013</strong>) 158e167 159<br />

1. Background<br />

It has become extensively clear over the past decade that millions<br />

of loci within the human genome can vary from person to<br />

person which affect gene and subsequent protein expression<br />

and influence our metabolism. Among our many happening<br />

metabolic processes, the action of drug is dependent on various<br />

metabolic processes that includes its activation and metabolism;<br />

thus determining its efficacy. For a drug like clopidogrel,<br />

a highly prescribed antiplatelet agent, a genetic testing<br />

which is able to predict variability in drug response has found<br />

its place in the field of pharmacogenomics.<br />

Clopidogrel, alone or combined with aspirin, is routinely<br />

used to treat patients with a variety of vascular disorders. It is<br />

a thienopyridine derivative; a prodrug activated in the liver by<br />

cytochrome P450 (CYP) enzymes mainly by CYP2C19. This<br />

active metabolite of the drug then binds irreversibly to the<br />

platelet adenosine diphosphate (ADP) receptor P2Y 12 which<br />

inhibits platelet degranulation, glycoprotein IIb/IIIa (GPIIbIIIa)<br />

receptor activation and thus platelet aggregation. 1e3 The<br />

pharmacodynamic response to clopidogrel varies widely from<br />

subject to subject, and about 25% of patients treated with<br />

standard clopidogrel doses display low ex vivo inhibition of<br />

ADP-induced platelet aggregation. Generally patients with<br />


160<br />

indian heart journal 65 (<strong>2013</strong>) 158e167<br />

prevalence of the polymorphisms. Of these subjects, blood<br />

sample was also collected of randomly selected 10 healthy<br />

individuals for platelet aggregation study.<br />

To analyze the effect of these polymorphism on platelet<br />

aggregation only those AMI cases (n ¼ 26) were selected and<br />

recruited who were not on clopidogrel before and were prescribed<br />

clopidogrel later. They were clopidogrel naïve patients<br />

screened from 100 AMI patients in the span of two years<br />

(2009e2011). The platelet aggregation of these patients was<br />

compared before and after the intake of the drug and thus<br />

platelet aggregation inhibition (PAI) was calculated and the<br />

effect of genotypes on PAI was analyzed. AMI patients were<br />

those who suffered an AMI with prolonged chest pain more<br />

than 30 min and ST segment elevation more than 0.1 mv on at<br />

least two adjacent ECG leads with elevated cardiac enzymes<br />

from ICCU ward. These AMI patients were with first episode of<br />

chest pain and had neither past history of such clinical<br />

symptoms nor were on any known medications for the same.<br />

AMI patients on prior clopidogrel treatment at the time of<br />

admission were excluded as their basal platelet aggregation<br />

could not be obtained which was essential to compare the<br />

extent of PAI after the intake of clopidogrel. Other exclusion<br />

criteria were valvular heart disease, known cardiomyopathy,<br />

malignancy, renal or liver diseases as well as subjects with<br />

systemic inflammatory disease to exclude any other complications<br />

and keep the patient population homogenous. These<br />

AMI patients at presentation were given 300 mg loading dose<br />

of clopidogrel and aspirin (150 mg) along with the standard<br />

treatment of nitrates antiarrhythmics, antifailures, statins<br />

and proton pump inhibitors (PPI) and subsequently were on<br />

maintenance dose of 75 mg clopidogrel per day. PPIs,<br />

frequently used in patients receiving clopidogrel and aspirin,<br />

are also metabolized by CYP2C19 and CYP3A4. 7 In our study,<br />

pantoprazole 40 was used as PPI for all the patients. In<br />

contrast to the reported negative omeprazole-clopidogrel drug<br />

interaction, the intake of pantoprazole or esomeprazole has<br />

been demonstrated, not to interfere with the activation of<br />

clopidogrel and was therefore not associated with impaired<br />

response. 26,27 The subjects were recruited in study only after<br />

obtaining informed consent. Information regarding their demographic<br />

status, clinical history, family history and medications<br />

were noted down in detail. The ethical committee of<br />

Sir H. N. Hospital and Research Centre and Rajawadi Municipal<br />

Hospital approved the study protocol. The analyzes of the<br />

samples were carried out at Sir H. N. Medical Research Society<br />

at Sir H. N. Hospital and Research Centre, Mumbai.<br />

2.2. Sample collection<br />

Blood samples of 102 healthy individuals were collected in the<br />

plain and EDTA anti-coagulated vacutainer after overnight,<br />

12 h fast for analyzing biochemical parameters and complete<br />

blood count respectively. Of these subjects, 10 healthy individuals<br />

were randomly selected and their blood sample was<br />

also collected in Na-Citrate vacutainer (3.2%) for platelet aggregation<br />

test. Peripheral blood samples of AMI patients were<br />

collected at presentation for routine analysis such as electrolytes,<br />

cardiac enzymes, complete blood count and prothrombin<br />

time. Remaining serum was stored at 80 C and<br />

EDTA blood sample at 4 C for further analysis. Peripheral<br />

blood of each AMI patient was collected in Na-Citrate vacutainer<br />

(3.2%) for platelet aggregation test at three different<br />

point of time e (1) baseline or 0 h e before clopidogrel loading<br />

dose (300 mg) administration, (2) 24 h after loading dose<br />

(300 mg) administration and before the next dose of 75 mg and<br />

(3) at 6 days when the patient was on 75 mg maintenance dose<br />

of clopidogrel.<br />

2.3. Platelet aggregation by turbidometric method<br />

Platelet aggregation test was carried out at the earlier mentioned<br />

three different point of time within 4 h of blood collection.<br />

Platelet rich plasma (PRP) and platelet poor plasma (PPP) were<br />

separated from each Na-citrated blood and then the test was<br />

carried out in duplicates using ADP (10 mM) as agonist in a two<br />

chambered Chronolog Platelet Aggregometer (model 490-2D).<br />

With the help of AggroLink software package, platelet aggregation<br />

was expressed as the maximal percent change in light<br />

transmittance from baseline in PRP with PPP used as a reference.<br />

All the patients were administered aspirin along with clopidogrel.<br />

The predominant antiplatelet effect of aspirin is mediated<br />

through its irreversible inactivation of cyclooxygenase (COX) 1,<br />

which is responsible for the formation of thromboxane A2, a<br />

potent vasoconstrictor and platelet aggregator, from arachidonic<br />

acid. 27 In the present study, final concentration of 10 mM<br />

ADP was chosen for platelet aggregation because at this concentration<br />

ADP induces complete platelet aggregation in a<br />

manner independent of thromboxane A2 production. 14,28,29<br />

2.4. Genotyping of polymorphisms<br />

DNA was extracted from peripheral leukocytes by modified<br />

Miller et al’s salting out procedure. 30 Genotyping was carried<br />

out by polymerase chain reaction (PCR) e based restriction<br />

enzyme digestion method for C3435T of MDR1 31 and i-T744C<br />

of P2Y 12 . 32 A multiplex PCR was standardized for CYP2C19*2<br />

and CYP2C19*3 using primer sequence of Pang et al 33 while<br />

CYP2C19*17 was genotyped using nested PCR. 34 The primers<br />

used for PCR of each polymorphism are given in Table 1. The<br />

Table 1 e Primers sequence.<br />

Gene<br />

Primer sequence<br />

MDR1 31<br />

(C3435T)<br />

CYP2C19*2 33<br />

(G681A)<br />

CYP2C19*3 33<br />

(G636A)<br />

CYP2C19*17 34<br />

(C806T)<br />

P2Y 12<br />

32<br />

(i-T744C)<br />

5 0 TGT TTT CAG CTG CTT GAT GG 3 0<br />

5 0 AAG GCA TGT ATG TTG GCC TC 3 0<br />

5 0 CAG AGC TTG GCA TAT TGT ATC 3 0<br />

5 0 GTA AAC ACA CAA CTA GTC AAT G 3 0<br />

5 0 AAA TTG TTT CCA ATC ATT TAG CT 3 0<br />

5 0 ACT TCA GGG CTT GGT CAA TA 3 0<br />

SET 1<br />

5 0 GCC CTT AGC ACC AAA TTC TC 3 0<br />

5 0 ATT TAA CCC CCT AAA AAA ACA CG 3 0<br />

SET 2<br />

5 0 AAA TTT GTG TCT TCT GTT CTC AAT G 3 0<br />

5 0 AGA CCC TGG GAG AAC AGG AC 3 0<br />

5 0 TCA CTT ATC TCT GGT GAA ATA AAA<br />

AGA TTA CGT A 3 0<br />

5 0 GTC AGA AAT GGC CTG TGT ATA TAT<br />

GGT CAT GAG 3 0


indian heart journal 65 (<strong>2013</strong>) 158e167 161<br />

content of the master mix and PCR conditions are depicted in<br />

Table 2. Taq polymerase and dNTPs were from FermentaseMBI.<br />

PCR was carried out in thermal cycler “T Gradient”<br />

from “Biometra” (Genetix Biotech Asia Pvt. Ltd.). Each sample<br />

was run in duplicates with a positive and negative control for<br />

each for a batch of samples. Amplified PCR product was then<br />

run on Agarose gel electrophoresis. Finally, the PCR products<br />

were subjected to restriction digestion with the enzymes Sau<br />

3A1, Sma I, Bam HI, Nsi I and Rsa I for identifying genotypes of<br />

C3435T of MDR1, CYP2C19*2, CYP2C19*3, CYP2C19*17 and<br />

i-T744C of P2Y 12 respectively. Fig. 1 shows the restriction<br />

digestion pattern of C3435T of MDR1, Fig. 2 shows CYP2C19*2<br />

and CYP2C19*3 genotypes of multiplex PCR, Figs. 3 and 4 show<br />

that of CYP2C19*17, and i-T744C of P2Y 12 , respectively on 10%<br />

polyacrylamide gel electrophoresis.<br />

in platelet aggregation between groups was assessed by<br />

ManneWhitney U test.<br />

3. Results<br />

3.1. Characteristics of the study population<br />

Demographic data and routine pathology data of 102 healthy<br />

individuals and 26 AMI patients is given in Table 3 and Table 4<br />

respectively. There was no significant difference in the age,<br />

body mass index and blood pressure amongst AMI patients.<br />

Six out of 26 patients were hypertensive while 4 were diabetic.<br />

3.2. Allelic frequencies<br />

2.5. Statistical analysis<br />

Genotype frequencies were estimated by the gene-counting<br />

method. Allelic frequencies were calculated from genotype<br />

frequencies. Genotypes were tested for deviations from HardyeWeinberg<br />

equilibrium. The change in platelet aggregation<br />

within a group from baseline to 24 h or 6th day was assessed<br />

using Wilcoxon Signed Rank test. The comparison of change<br />

The genotype and allele frequencies for all the polymorphisms<br />

studied are given in Table 5 and Table 6 for<br />

healthy individuals and AMI patients, respectively. The study<br />

population did not show the presence of the variant allele of<br />

CYP2C19*3. The observed genotype distributions did not<br />

deviate from HardyeWeinberg equilibrium.<br />

As the sample size of healthy individuals (n ¼ 102) and that<br />

of AMI patients (n ¼ 26) differed, the significance of difference<br />

Table 2 e Details of the PCR protocol for genotyping polymorphisms.<br />

Genotype Content of the PCR reaction PCR conditions PCR amplified<br />

product<br />

MDR1 (C3435T)<br />

1 U Taq DNA polymerase, 2.5 ml of 10X<br />

Buffer with 25 mM MgCl 2 , 0.5 ml of10mM<br />

dNTPs, 10 pmoles of primers and 100 ng<br />

of genomic DNA<br />

Initial denaturation at 95 C for 5 min<br />

followed by 30 cycles of denaturation<br />

at 95 C for 45 s, annealing at 62 C<br />

for 30 s and extension at 72 C for<br />

30 s followed by final extension at<br />

72 C for 5 min.<br />

The PCR amplification conditions<br />

consisted 3 steps e an initial<br />

denaturation step at 95 C for 5 min<br />

followed by 30 cycles of denaturation<br />

at 95 C for 45 s, annealing at 58 C<br />

for 50 s and extension at 72 C for 1<br />

min followed by final extension at<br />

72 C for 3 min<br />

An initial denaturation step at 95 C<br />

for 5 min followed by 30 cycles of<br />

denaturation 95 C for 45 s, annealing<br />

at 62 C for 30 s and denaturation at<br />

72 C for 30 s followed by final<br />

extension at 72 C for 5 min.<br />

Second PCR<br />

Initial denaturation step at 95 C for 5<br />

min followed by 30 cycles of<br />

denaturation at 95 C for 45 s,<br />

annealing at 62 C for 30 s and<br />

extension at 72 C for 30 s followed<br />

by final extension at 72 C for 3 min<br />

Initial denaturation step at 95 C for 5<br />

min followed by 30 cycles of<br />

denaturation at 95 C for 45 s,<br />

annealing at 60 C for 2 min and<br />

extension at 72 C for 2 min followed<br />

by final extension at 72 C for 5 min<br />

197 base pair (bp)<br />

CYP2C19*2 and<br />

CYP2C19*3<br />

polymorphisms<br />

by multiplex PCR<br />

The 50 ml reaction mixture contained 2 U<br />

Taq DNA polymerase, 5 ml 10X Buffer with<br />

25 mM MgCl 2 ,1ml 10 mM dNTPs and 15<br />

pmoles of each specific primers and 100 ng<br />

of genomic DNA<br />

321 bp of CYP2C19*2<br />

(G681A) and 271 bp of<br />

CYP2C19*3 (G636A)<br />

CYP2C19*17<br />

polymorphism<br />

by nested PCR<br />

First PCR<br />

The first 25 ml reaction mixture contained 1 U<br />

Taq DNA polymerase, 2.5 ml of 10X buffer<br />

with 25 mM MgCl 2 , 0.5 ml of 10 mM dNTPs<br />

and 10 pmoles of each specific primers and<br />

100 ng of genomic DNA<br />

Second PCR<br />

The next 50 ml reaction mixture contained 1U<br />

Taq DNA polymerase, 5 ml of 10X Buffer with<br />

25 mM MgCl 2 , 0.5 ml of 10 mM dNTPs and 10<br />

pmoles of each specific primers. For each<br />

sample to be genotyped 1 ml of earlier amplified<br />

PCR product was added later.<br />

2 U Taq DNA polymerase (Fermentas - MBI),<br />

2.5 ml 10X Buffer with 25 mM MgCl 2 , 0.5 ml of<br />

10 mM dNTPs and 10 pmoles of each specific<br />

primers and 100 ng of genomic DNA.<br />

473bp<br />

143bp<br />

P2Y 12<br />

(i-T744C)<br />

220bp


162<br />

indian heart journal 65 (<strong>2013</strong>) 158e167<br />

Fig. 1 e The restriction digestion pattern of MDR1 [C3435T]<br />

on 10% polyacrylamide gel electrophoresis. Lane 1: PCR<br />

product of 197 bp. Lane 2 and 4: heterozygous genotype<br />

(C3435T) showing two bands of 197 bp and 158 bp. Lane 3:<br />

O 0 Range Ruler, 20 bp, ready to use DNA Ladder from MBI<br />

Fermentas. Lane 4: homozygous mutant genotype<br />

(T3435T) showing one band of 158 bp. Lane 6: wild type<br />

genotype (C3435C) showing one band of 197 bp.<br />

Fig. 3 e The restriction digestion pattern of CYP2C19*17 on<br />

10% polyacrylamide gel electrophoresis. Lane 1: PCR<br />

products of 143 bp. Lane 2: wild type genotype (*1 3 *1)<br />

showing one band of 116 bp. Lane 3: Heterozygous<br />

genotype (*1 3 *17) showing two bands 143 bp and 116 bp.<br />

Lane 4 to 6: homozygous mutant genotype (*17 3 *17)<br />

showing one band of 143 bp. Lane 7: O 0 Range Ruler, 20 bp,<br />

ready to use DNA Ladder from MBI Fermentas.<br />

of prevalence of the genotypes and alleles (using Chi Square<br />

statistics with Yates Corrections) was not calculated. However,<br />

it was observed that the prevalence of wild type allele<br />

frequency of CYP2C19*17 (C806, 0.897 > 0.865) and the variant<br />

allele frequency of 3435T of MDR1 (0.576 > 0.524), CYP2C19*2<br />

(681A, 0.423 > 0.352) as well as i744C of P2Y 12 (0.173 > 0.088) of<br />

AMI patients although marginal, were more as compared that<br />

of healthy individuals (Tables 5 and 6).<br />

3.3. Platelet aggregation<br />

The platelet aggregation of healthy individuals (n ¼ 10) was in<br />

the range of 60e80% (70.4 8.87) almost similar to average<br />

Fig. 2 e The restriction digestion pattern of CYP2C19*2 and<br />

CYP2C19*3 on 10% polyacrylamide gel electrophoresis.<br />

Lane 1: PCR products of 321 bp and 271 bp for CYP2C19*2<br />

and CYP2C19*3 respectively. Lane 2eLane 4: wild type<br />

genotype (*1 3 *1) of CYP2C19*2 showing two bands 212 bp<br />

and 109 bp and wild type genotype (*1 3 *1) of CYP2C19*3<br />

showing two bands 175 bp and 196 bp. Lane 3:<br />

Heterozygous genotype (*1 3 *2) of CYP2C19*2 showing<br />

three bands 321 bp, 212 bp and 109 bp and wild type<br />

genotype (*1 3 *1) of CYP2C19*3 showing two bands 175 bp<br />

and 196 bp. Lane 4: homozygous mutant genotype (*2 3 *2)<br />

showing one band of 321 bp and wild type genotype<br />

(*1 3 *1) of CYP2C19*3 showing two bands 175 bp and 196<br />

bp. Lane 5: O 0 Range Ruler, 20 bp, ready to use DNA Ladder<br />

from MBI Fermentas.<br />

Fig. 4 e The restriction digestion pattern of P2Y 12 [i-T744C]<br />

on 10% polyacrylamide gel electrophoresis. Lane 1: PCR<br />

products of 220 bp. Lane 2: wild type genotype (i-T744T)<br />

showing one band of 220 bp. Lane 3: heterozygous<br />

genotype (i-T744C) showing two bands 220 bp and 196 bp.<br />

Lane 4: Homozygous mutant genotype (i-C744C) showing<br />

one band of 196 bp. Lane 5: O 0 Range Ruler, 20 bp, ready to<br />

use DNA Ladder from MBI Fermentas.


indian heart journal 65 (<strong>2013</strong>) 158e167 163<br />

Table 3 e Demographic data.<br />

Parameters<br />

Healthy individuals<br />

(n ¼ 102)<br />

AMI patients<br />

(n ¼ 26)<br />

M:F 47: 55 19:7<br />

Age [years] 43.59 10.74 51 10<br />

BMI [kg/m 2 ] 24.4 4.53 23.98 2.12<br />

SBP [mmHg] 118 9.23 135 29<br />

DBP [mmHg] 78.96 6.73 82 17<br />

Smoking 14 14<br />

Alcohol consumption 15 8<br />

Hypertension e 6<br />

Diabetes mellitus e 4<br />

baseline platelet aggregation (0 h) of 26 AMI patients (69.16%).<br />

The platelet aggregation of all AMI patients (n ¼ 26) dropped to<br />

45.6% at 24 h subsequent to the administration of 300 mg<br />

loading dose with PAI of only 23.56%. Further, platelet aggregation<br />

dropped to 43.4% at 6 days on further 75 mg maintenance<br />

dose per day of clopidogrel demonstrating PAI of only<br />

25.76%.<br />

Among 26 AMI patients, none demonstrated homozygous<br />

mutant (T806T) genotype of CYP2C19*17. All the patients were<br />

associated with either wild type or heterozygous genotypes<br />

associated with decreased enzyme function. Also there were<br />

only 4 patients each with wild type (C3435C) genotype of MDR1<br />

and rests were with variant genotypes associated decrease<br />

intestinal absorption. Therefore, further analysis of effect of<br />

polymorphisms on response to clopidogrel was carried out<br />

with respect to CYP2C19*2 genotypes and patients were<br />

grouped according to common genotypes as depicted in<br />

Table 7. Group I, II and III were with wild type (G681G), heterozygous<br />

(G681A) and homozygous mutant (A681A) genotype<br />

of CYP2C19*2 respectively and wild type genotype (i-T744T) of<br />

P2Y 12 . Subsequently, subjects were grouped as per variant<br />

genotypes of i-T744C of P2Y 12. Group IV was one case having<br />

homozygous mutant genotype (A681A) of CYP2C19*2 and<br />

heterozygous genotype (i-T744C) of P2Y 12 while three cases<br />

with heterozygous genotype (G681A) of CYP2C19*2 and homozygous<br />

mutant genotype (i-C744C) of P2Y 12 were included<br />

Table 4 e Routine pathology data.<br />

Parameters<br />

Healthy<br />

individuals<br />

(n ¼ 102)<br />

AMI patients<br />

(n ¼ 26)<br />

Total cholesterol (TC) (mg/dl) 188.5 35.38 172.5 44.4<br />

HDL-cholesterol (HDL-C) (mg/dl) 44.63 9.29 38.4 7.65<br />

TC/HDL-C 4.31 0.86 4.47 0.74<br />

LDL cholesterol (LDL-C) (mg/dl) 124.8 25.5 105.9 40.1<br />

LDL-C/HDL-C 2.8 0.71 2.8 0.6<br />

Triglycerides (mg/dl) 102.6 41.5 127.4 61.02<br />

VLDL cholesterol (mg/dl) 22.24 13.11 25.5 12.2<br />

Glucose (mg/dl) 95.1 10.1 154.2 68.3<br />

SGOT U/L 16 3.25 51.4 30.12<br />

SGPT U/L 20.1 2.3 38.2 15.8<br />

BUN (mg/dl) 8.1 2.37 10.1 7.32<br />

Creatinine (mg/dl) 0.7 0.12 0.98 0.24<br />

Uric acid (mg/dl) 4.0 1.3 4.95 1.84<br />

Total WBC count/mL 6975 1602 11 210 2926<br />

Platelet count 10 3 /mL 259 109.5 328 137<br />

in Group V. There was only one case with wild type genotype<br />

(G681G) of CYP2C19*2 and homozygous mutant genotype<br />

(i-C744C) of P2Y 12 (Group VI). The PAI at 24 h was in the<br />

decreasing order; 37%, 22%, 18.5%, 18.0%, 14.3% and 11.0% for<br />

Group I, II, III, IV, V and VI respectively.<br />

Statistical analysis of Group I and Group II with 6 and 13<br />

cases, respectively demonstrated that decrease in platelet<br />

aggregation i.e. PAI of Group I from 0 h to 24 h (37%, p ¼ 0.002)<br />

and 0 to 6th day (27%, p ¼ 0.027) was significant. The same was<br />

also observed for Group II for PAI from baseline to 24 h (22.0%,<br />

p ¼ 0.001) as well as from baseline to 6th day (24.3%, p ¼ 0.001).<br />

PAI at 24 h of Group II (22.0%) was less by 15% as compared to<br />

Group I (37.0%) but this decrease in PAI did not reach statistical<br />

significance of p< 0.05 (p ¼ 0.072). PAI at 6 days from baseline<br />

of Group I and II were almost similar 27% and 24.3%, respectively<br />

and did not reach statistical significance of p < 0.05<br />

(p ¼ 0.639). There were 3 hypertensives and one diabetic patient<br />

in Group I while in Group II there were 2 hypertensives<br />

and 2 diabetic patients. After excluding these patients, the<br />

difference in the PAI between Group I and II was 16.0% almost<br />

similar to that obtained on including these patients (15.0%).<br />

There was no significant difference in the BMI or age between<br />

the two patient groups (data not shown).<br />

Table 7 also depicts the CYP2C19*17 and C3435T genotypes<br />

of MDR1. In Group I, out of 6 patients, 5 had wild type genotype<br />

(C806C) for CYP2C19*17 while just one showed heterozygous<br />

genotype (C806T) for the same. Whereas in Group II, 11 out of<br />

13 had wild type genotype for CYP2C19*17 and just 2 showed<br />

heterozygous genotype for the same. On the other hand, in<br />

Group I out of 6 patients 2 were heterozygous (C3435T) and 4<br />

were homozygous mutant (T3435T) thus all were with variant<br />

genotypes of C3435T polymorphism of MDR1. In Group II, out<br />

of 13 patients, 7 were heterozygous and 3 each showed homozygous<br />

mutant and wild type genotype (C3435C) for C3435T<br />

polymorphism of MDR1.<br />

Remaining 7 patients could not be included in Group I or II<br />

and therefore in Table 7 they are grouped separately. Their PAI<br />

at 24 h was less as compared to Group I. Of these notably, 3<br />

patients of Group V with not only heterozygous genotype for<br />

CYP2C19*2 but also homozygous mutant genotype (i-C744C) of<br />

P2Y 12 demonstrated further (22.0%e14.3%) 7.7% less PAI as<br />

compared to Group II at 24 h. One patient with wild type<br />

genotype of CYP2C19*2 and homozygous mutant genotype<br />

(i-C744C) of P2Y 12 demonstrated only 11% PAI at 24 h. At 6th<br />

day, the PAI for all seven patients was either similar or near to<br />

Group I patients.<br />

4. Discussion<br />

The present study demonstrated the prevalence of polymorphisms<br />

of drug-efflux transporter protein responsible for<br />

drug absorption (MDR1), one of the metabolizing enzymes of<br />

clopidogrel (CYP2C19) and its target (P2Y 12 ), in our study<br />

population. Its effect was demonstrated on antiplatelet activity<br />

of clopidogrel in AMI patients assessed ex vivo by<br />

ADP-induced platelet aggregation.<br />

In our study population variant allele frequency of 3435T of<br />

MDR1 affecting intestinal absorption was found to be 52.4%<br />

(0.524) nearing to that reported for <strong>Indian</strong> population


164<br />

indian heart journal 65 (<strong>2013</strong>) 158e167<br />

Table 5 e Genotype and allele frequencies of healthy individuals (n [ 102) for all the polymorphisms studied.<br />

Genotype frequency<br />

Allele frequency<br />

Wild type Heterozygous Homozygous Wild type allele Mutant allele<br />

MDR1<br />

(C3435T)<br />

CYP2C19*2<br />

(G681A)<br />

CYP2C19*3<br />

(G636A)<br />

CYP2C19*17<br />

(C806T)<br />

P2Y 12<br />

(i-T744C)<br />

26.4% (27) 42.1% (43) 31.4% (32) 3435C ¼ 0.475 3435T ¼ 0.524<br />

46% (47) 37% (38) 16.7% (17) 681G ¼ 0.647 681A ¼ 0.352<br />

100% (102) 0 0 636G ¼ 1.0 e<br />

81.4% (83) 16.7% (17) 1.96% (2) 806C ¼ 0.897 806T ¼ 0.102<br />

85.2% (87) 11.8% (12) 2.94% (3) i744T ¼ 0.911 i744C ¼ 0.088<br />

(0.61e0.62). 21,22 In African, 35 Asian and Caucasian 36 population,<br />

allele frequency of 3435T of MDR1 has been reported to be<br />

21.0%, 42.0% and 55.0%, respectively. The variant allele frequency<br />

of CYP2C19*2 (681A) associated with poor metabolizer<br />

type, was observed to be 35.2% (0.352) in the present study<br />

which is similar to that reported by Adithan et al 23 for<br />

Tamilian population (37.9%) and Mizutani et al 36 who have<br />

reported 30e35% in Asians and is greater than reported for<br />

African (17e20%) 35 and Caucasians (13e18%). 36 In the present<br />

study, the presence of CYP2C19*3 variant allele (636A), also<br />

associated with poor metabolizer type, was not observed<br />

which has also been reported by Pang et al 33 in Malaysian<br />

subjects. However, Adithan et al 22 have reported CYP2C9*3<br />

variant allele frequency as 2.2% in Tamilian population. In<br />

African, 35 Asians and Caucasians, 36 the variant allele frequency<br />

of CYP2C19*3 has been reported to be


indian heart journal 65 (<strong>2013</strong>) 158e167 165<br />

Table 7 e Platelet aggregation, platelet aggregation inhibition (PAI) of AMI patients (n [ 26) grouped as per genotypes.<br />

Groups<br />

Platelet<br />

Platelet aggregation<br />

Genotypes<br />

aggregation (%)<br />

inhibition (PAI) (%)<br />

CYP2C19 P2Y 12 CYP2C19 MDR 1<br />

0 h 24 h 6 days At 0 h At 6 days *2 *3 -i T744C *17 C3435T<br />

Group I (n ¼ 6) 67.8 30.7 40.3 37 27.3 W W W W HMM<br />

W W W W HMM<br />

W W W W HMM<br />

W W W W HMM<br />

W W W W HZ<br />

W W W HZ HZ<br />

Group II (n ¼ 13) 67.15 45.15 42.15 22 24 HZ W W W HZ<br />

HZ W W W HZ<br />

HZ W W W HZ<br />

HZ W W W HMM<br />

HZ W W W HZ<br />

HZ W W HZ W<br />

HZ W W W HMM<br />

HZ W W W W<br />

HZ W W W HMM<br />

HZ W W HZ HZ<br />

HZ W W W W<br />

HZ W W W HZ<br />

HZ W W W HZ<br />

Group III (n ¼ 2) 71.5 53 51 18.5 20.5 HMM W W HZ HZ<br />

HMM W W HZ HZ<br />

Group IV (n ¼ 1) 79 61 47 18 32 HMM W HZ W HZ<br />

Group V (n ¼ 3) 76.7 62.3 50 14.3 26.7 HZ W HMM HZ HMM<br />

HZ W HMM HZ HZ<br />

HZ W HMM W W<br />

Group VI (n ¼ 1) 64 53 33 11 31 W W HMM W HZ<br />

Wewild type genotype, HZeheterozygous genotype, HMMehomozygous mutant genotype.<br />

were on maintenance dose of 75 mg clopidogrel per day,<br />

indicating that a part of low pharmacodynamic response of<br />

clopidogrel due to the CYP2C19*2 polymorphism may be<br />

reversed to some extent by repeated dosing.<br />

Fontana et al 20 have identified a P2Y 12 receptor haplotype<br />

H2 to be strongly associated with an increase in ADP-induced<br />

platelet aggregation. In 2008, Malek et al 32 reported that the<br />

co-existence of the variant i-744C allele of P2Y 12 and the<br />

variant 681A allele of CYP2C19*2 is associated with persisting<br />

platelet activity in patients with acute coronary syndrome<br />

(ACS) on clopidogrel treatment. Similar to this finding, in the<br />

present study, there were three patients with homozygous<br />

mutant genotype (i-C744C) for P2Y 12 receptor in combination<br />

with heterozygous genotype for CYP2C19*2 (Group V) who<br />

showed additional 7.7% decreased PAI as compared to patients<br />

of Group II with wild type genotype (i-T744T) of P2Y 12<br />

receptor and heterozygous genotype (G681A) of CYP2C19*2.<br />

Further Malek et al 32 have reported that the carriers of i-744C<br />

allele of P2Y 12 without variant allele of CYP2C19*2 did not have<br />

significantly decreased platelet response to ADP suggesting<br />

that i-T744C polymorphism of P2Y 12 could also independently<br />

elevate platelet reactivity. In the present study we had only<br />

one case with homozygous mutant (i-C744C) genotype of<br />

P2Y 12 and wild type of CYP2C19*2 which was also associated<br />

with increased response to ADP with PAI of only 11% at 24 h.<br />

Small study population for analyzing the platelet aggregation<br />

effect is the major limitation of our study which was<br />

further affected by variations in genotypes. To see the extent<br />

of clopidogrel to inhibit the platelet aggregation, it was very<br />

important that these patients were without any prior antiplatelet<br />

intake. For this reason, patients with prior antiplatelet<br />

treatment were excluded and these 26 AMI clopidogrel naïve<br />

patients were screened from 100 AMI patients in the span of<br />

two years from January 2009 to <strong>Mar</strong>ch 2011.<br />

5. Conclusion<br />

In our study the occurrence of variant alleles of MDR1 (3435T),<br />

CYP2C19*2 (681A), P2Y 12 (i-744C) as well as wild type allele of<br />

CYP2C19*17 (C806) associated with decreased clopidogrel<br />

response were observed. The present study did show a trend<br />

toward impaired response of clopidogrel to inhibit platelet<br />

aggregation with variant genotypes of CYP2C19*2 and iT744C<br />

of P2Y 12 compared to respective wild type genotype at 24 h.<br />

Similar studies of larger sample size and of longer follow-up<br />

may strengthen our view for individualized antiplatelet<br />

treatment based on genotypic analysis for patients with ACS<br />

and those undergoing PCI who receive loading dose of 300 mg<br />

of clopidogrel.<br />

Funding source<br />

The work is being supported by the grants from Sir H. N.<br />

Medical Research Society.


166<br />

indian heart journal 65 (<strong>2013</strong>) 158e167<br />

Conflicts of interest<br />

All authors have none to declare.<br />

Acknowledgment<br />

Authors would like to acknowledge Sir H. N. Hospital and<br />

Research Centre, and Rajawadi Municipal Hospital for<br />

recruitment of the patients and Sir H. N. Medical Research<br />

Society for the financial support.<br />

references<br />

1. Yusuf S, Zhao F, Mehta SR, et al. Effects of Clopidogrel in<br />

addition to aspirin in patients with acute coronary syndromes<br />

without ST-segment elevation. N Engl J Med.<br />

2001;345:494e502.<br />

2. Sabatine MS, Cannon CP, Gibson CM, et al. Addition of<br />

clopidogrel to aspirin and fibrinolytic therapy for myocardial<br />

infarction with ST-segment elevation. N Engl J Med.<br />

2005;352:1179e1189.<br />

3. King 3rd SB, Smith Jr SC, Hirshfeld Jr JW, et al. 2007 focused<br />

update of the ACC/AHA/SCAI 2005 Guideline Update for<br />

Percutaneous Coronary Intervention: a report of the<br />

American College of Cardiology/American <strong>Heart</strong> Association<br />

Task Force on Practice Guidelines: 2007 Writing Group to<br />

Review New Evidence and Update the ACC/AHA/SCAI 2005<br />

Guideline Update for Percutaneous Coronary Intervention,<br />

Writing on behalf of the 2005 Writing Committee. Circulation.<br />

2008;117:261e295.<br />

4. Gurbel PA, Bliden KP, Hiatt BL, O’Connor CM. Clopidogrel for<br />

coronary stenting: response variability, drug resistance, and<br />

the effect of pretreatment platelet reactivity. Circulation.<br />

2003;107:2908e2913.<br />

5. Muller I, Besta F, Schulz C, Massberg S, Schomig A, Gawaz M.<br />

Prevalence of Clopidogrel non-responders among patients<br />

with stable angina pectoris scheduled for elective coronary<br />

stent placement. Thromb Haemost. 2003;89:783e787.<br />

6. Nguyen TA, Diodati JG, Pharand C. Resistance to clopidogrel: a<br />

review of the evidence. J Am Coll Cardiol. 2005;45:1157e1164.<br />

7. Wiviott SD, Antman EM. Clopidogrel resistance: a new chapter<br />

in a fast-moving story. Circulation. 2004;109:3064e3067.<br />

8. Taubert D, von Beckerath N, Grimberg G, et al. Impact of<br />

P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther.<br />

2006;80:486e501.<br />

9. Fung KL, Gottesman MM. A synonymous polymorphism in a<br />

common MDR1 (ABCB1) haplotype shapes protein function.<br />

(BBA)dProteins and Proteomics. 2009;1794:860e871.<br />

10. Tang K, Wong LP, Lee EJD, Chong SS, Lee CGL. Genomic<br />

evidence for recent positive selection at the human MDR1<br />

gene locus. Hum Mol Genet. 2004;13:783e797.<br />

11. Kazui M, Nishiya Y, Ishizuka T, et al. Identification of the<br />

human cytochrome P450 enzymes involved in the two<br />

oxidative steps in the bioactivation of clopidogrel to its<br />

pharmacologically active metabolite. Drug Metab Dispos.<br />

2010;38:92e99.<br />

12. De Morais SM, Wilkinson GR, Blaisdell J, Meyer UA,<br />

Nakamura K, Goldstein JA. Identification of a new genetic<br />

defect responsible for the polymorphism of (S)-mephenytoin<br />

metabolism in Japanese. Mol Pharmacol. 1994;46:594e598.<br />

13. De Morais SM, Wilkinson GR, Blaisdell J, Nakamura K,<br />

Meyer UA, Goldstein JA. The major genetic defect responsible<br />

for the polymorphism of S-mephenytoin metabolism in<br />

humans. J Biol Chem. 1994;269:15419e15422.<br />

14. Hulot JS, Bura A, Villard E, et al. Cytochrome P450 2C19 lossof-function<br />

polymorphism is a major determinant of<br />

clopidogrel responsiveness in healthy subjects. Blood.<br />

2006;108:2244e2247.<br />

15. Brandt JT, Close SL, Iturria SJ, et al. Common polymorphisms<br />

of CYP2C19 and CYP2C9 affect the pharmacokinetic and<br />

pharmacodynamic response to clopidogrel but not prasugrel.<br />

J Thromb Haemost. 2007;5:2429e2436.<br />

16. Trenk D, Hochholzer W, Fromm MF, et al. Cytochrome P450<br />

2C19 681G>A polymorphism and high on-Clopidogrel platelet<br />

reactivity associated with adverse 1-year clinical outcome of<br />

elective percutaneous coronary intervention with drug<br />

eluting or bare-metal stents. J Am Coll Cardiol.<br />

2008;51:1925e1934.<br />

17. Sim SC, Risinger C, Dahl ML, et al. A common novel CYP2C19<br />

gene variant causes ultrarapid drug metabolism relevant for<br />

the drug response to proton pump inhibitors and<br />

antidepressants. Clin Pharmacol Ther. 2006;79:103e113.<br />

18. Sibbing D, Koch W, Gebhard D, et al. Cytochrome 2C19*17<br />

allelic variant, platelet aggregation, bleeding events, and<br />

stent thrombosis in clopidogrel-treated patients with<br />

coronary stent placement. Circulation. 2010;121:512e518.<br />

19. Hollopeter G, Jantzen HM, Vincent D, et al. Identification of<br />

the platelet ADP receptor targeted by antithrombotic drugs.<br />

Nature. 2001;409:202e207.<br />

20. Fontana P, Dupont A, Gandrille S, et al. Adenosine<br />

diphosphate e induced platelet aggregation is associated<br />

with P2Y12 gene sequence variations in healthy subjects.<br />

Circulation. 2003;108:989e995.<br />

21. Balram C, Sharma A, Sivathasan C, Lee EJ. Frequency of<br />

C3435T single nucleotide MDR1 genetic polymorphism in an<br />

Asian population: phenotypicegenotypic correlates. Br J Clin<br />

Pharmacol. 2003;56:78e83.<br />

22. Ashavaid T, Raje H, Shalia K, Shah B. Effect of gene<br />

polymorphisms on the levels of calcineurin inhibitors in<br />

<strong>Indian</strong> renal transplant recipients. <strong>Indian</strong> J Nephrol.<br />

2010;20:146e151.<br />

23. Adithan C, Gerard N, Vasu S, Rosemary J, Shashindran CH,<br />

Krishnamoorthy R. Allele and genotype frequency of CYP2C19<br />

in a Tamilian population. Br J Clin Pharmacol. 2003;56:331e333.<br />

24. Panchabhai TS, Noronha SF, Davis S, Shinde VM,<br />

Kshirsagar NA, Gogtay J. Evaluation of the activity of CYP2C19<br />

in Gujrati and <strong>Mar</strong>wadi subjects living in Mumbai (Bombay).<br />

BMC Clin Pharmacol. 2006;24:6e8.<br />

25. Ellis KJ, Souffer GA, McLeod HL, Lee CR. Clopidogrel<br />

pharmacogenomics and risk of inadequate platelet<br />

inhibition: US FDA recommendations. Pharmacogenomics.<br />

2009;10:1799e1817.<br />

26. Siller-Matula JM, Spiel AO, Lang IM, Kreiner G, Christ G,<br />

Jilma B. Effects of pantoprazole and esomeprazole on<br />

platelet inhibition by clopidogrel. Am <strong>Heart</strong> J.<br />

2009;157:148e1e148e5.<br />

27. Cattaneo M. Aspirin and clopidogrel: efficacy, safety and the<br />

issue of drug resistance. Arterioscler Thromb Vasc Biol.<br />

2004;24:1980e1987.<br />

28. Dörr G, Schmidt G, Gräfe M, Regitz-Zagrosek V, Fleck E. Effects<br />

of combined therapy with clopidogrel and acetylsalicylic acid<br />

on platelet glycoprotein expression and aggregation. J<br />

Cardiovasc Pharmacol. 2002;39:523e532.<br />

29. Wilkinson GR. Drug metabolism and variability among<br />

patients in drug response. N Engl J Med. 2005;352:2211e2221.<br />

30. Miller SA, Dykes DD, Polesky HF. A simple salting out<br />

procedure for extracting DNA from human nucleated cells.<br />

Nucleic Acids Res. 1988;16:1215.


indian heart journal 65 (<strong>2013</strong>) 158e167 167<br />

31. Cizmarikova M, Wagnerova M, Schonova L, Habalova V,<br />

Kohut A, Linkova A. MDR1 (C3435T) polymorphism: relation<br />

to the risk of breast cancer and therapeutic outcome.<br />

Pharmacogenomic J. 2010;10:62e69.<br />

32. Malek LA, Kisiel B, Spiewak M, et al. Coexisting<br />

polymorphisms of P2Y12 and CYP2C19 genes as a risk factor<br />

for persistent platelet activation with clopidogrel. Circ J.<br />

2008;72:1165e1169.<br />

33. Pang YS, Wong LP, Lee TC, Mustafa AM, Mohamed Z, Lang CC.<br />

Genetic polymorphism of Cytochrome P450 2C19 in healthy<br />

Malaysian subjects. Br J Clin Pharmacol. 2004;58:332e335.<br />

34. Baldwin RM, Ohlsson S, Pedersen RS, et al. Increased<br />

omeprazole metabolism in carriers of the CYP2C19*17 allele; a<br />

pharmacokinetic study in healthy volunteers. Br J Clin<br />

Pharmacol. 2008;65:767e774.<br />

35. Xie H-G, Kim RB, Wood AJJ, Stein CM. Molecular basis of<br />

ethnic differences in drug disposition and response. Annu Rev<br />

Pharmacol Toxicol. 2001;41:815e850.<br />

36. Mizutani T. PM frequencies of major CYPs in Asians and<br />

Caucasians. Drug Metabol Rev. 2003;35(2e3):99e106.<br />

37. Giusti B, Gori AM, <strong>Mar</strong>cucci R, et al. Cytochrome P450 2C19<br />

loss-of-function polymorphism, but not CYP3A4 IVS10 þ 12G/<br />

A and P2Y12 T744C polymorphisms, is associated with<br />

response variability to dual antiplatelet treatment in highrisk<br />

vascular patients. Pharmacogenet Genomics.<br />

2007;17:1057e1064.


indian heart journal 65 (<strong>2013</strong>) 180e186<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Original Article<br />

Association between erectile dysfunction and coronary artery<br />

disease and it’s severity<br />

A. Sai Ravi Shanker a, *, B. Phanikrishna b , C. Bhaktha Vatsala Reddy c<br />

a Cardiologist, Department of Cardiology, Narayana Medical College, Chinta Reddy Palem, Nellore 524003, Andhra Pradesh, India<br />

b Associate Professor, Narayana Medical College, Chinta Reddy Palem, Nellore 524003, Andhra Pradesh, India<br />

c Assistant Professor, Narayana Medical College, Chinta Reddy Palem, Nellore 524003, Andhra Pradesh, India<br />

article info<br />

Article history:<br />

Received 31 July 2012<br />

Accepted 16 February <strong>2013</strong><br />

Available online 24 February <strong>2013</strong><br />

Keywords:<br />

Erectile dysfunction<br />

Coronary artery disease<br />

Acute coronary syndrom<br />

Gensini’s score<br />

International Index of Erectile<br />

dysfunction<br />

abstract<br />

Background/aims: To investigate the prevalence of erectile dysfunction (ED) in patients with<br />

coronary artery disease (CAD), its relationship between the severity of ED and the extent of<br />

coronary vessel involvement and to register the mean time interval between them.<br />

Methods: 240 patients with CAD divided into three age-matched groups: Group 1 (n ¼ 60),<br />

ACS with one-vessel disease (1VD); group 2 (n ¼ 60), ACS with 2,3VD; group 3 (n ¼ 60), CSA.<br />

Control group (C, n ¼ 60) was composed of patients with suspected CAD who were found to<br />

have entirely normal coronary arteries by angiography. ED as any value


indian heart journal 65 (<strong>2013</strong>) 180e186 181<br />

endothelial dysfunction, leading to restrictions in blood<br />

flow. 5,6 Prevalence of ED as high as 75% has been reported in<br />

the established CAD patients. 7e12<br />

Atherosclerosis can play a major role in the development<br />

of ED both in the general population and in diabetic<br />

patients. 13e17 In the diabetic population, the prevalence of<br />

silent CAD is particularly high. 18,19<br />

Evidence to support ED as a predictor of CAD is:<br />

A significant proportion of men with ED exhibit early signs<br />

of CAD.<br />

Men with pre-existing ED may develop more severe CAD<br />

than those without ED.<br />

The interval between the onset of ED symptoms and the<br />

occurrence of CAD symptoms is estimated at 2e3 years and<br />

a cardiovascular event at 3e5 years.<br />

There is a common endothelial pathology underlying both<br />

ED and CAD.<br />

Erectile dysfunction is associated with increased all-cause<br />

mortality primarily through its association with CAD<br />

mortality.<br />

Erectile dysfunction is associated with significant changes<br />

in established cardiovascular risk factors such as fasting<br />

lipids, fasting glucose, body mass index (BMI), C-reactive<br />

protein (CRP) and homocysteine. 20e23 Men with ED generally<br />

exhibit more severe CAD and left ventricular dysfunction than<br />

those without ED, 24e26 and the severity of ED may also be<br />

correlated with the severity of CAD. 27 It should be noted,<br />

however, that penile Doppler testing cannot be reliably used<br />

to identify at-risk men because of its average sensitivity and<br />

specificity, low positive predictive value and high negative<br />

predictive value. 28 In around two-thirds of men, the onset of<br />

CAD is preceded by ED (Montorsi et al.). A number of studies<br />

have estimated the interval between the onset of ED symptoms<br />

and the occurrence of CAD symptoms as 2e3 years and a<br />

cardiovascular event [myocardial infarction (MI) or stroke] as<br />

3e5 years, 29,30 although longer time frames have been<br />

reported. 31<br />

Using Framingham risk scores, the relative risk of developing<br />

CAD within 10 years in men with moderate-severe ED<br />

has been estimated as 4.9% in those aged 30e39 years,<br />

increasing to 21.1% in those aged 60e69 years. 32 This compares<br />

with 4.3% and 16.6% in men without ED for the same age<br />

groups, i.e. an increase in relative risk of 1.14 and 1.27<br />

respectively. The risk of experiencing a cardiovascular event<br />

within a 10-year timeframe is increased by 1.3e1.6 times in<br />

men with ED vs. men without ED. 33,34<br />

2. Aims and objectives of the study<br />

To investigate the prevalence of ED in patients with CAD and<br />

to evaluate the relationship between the severity of ED and<br />

the extent of coronary vessel involvement and to register the<br />

first symptom and the mean time interval between them.<br />

We tested the hypothesis that ED prevalence is related to<br />

coronary atherosclerotic burden that in turn is related to the<br />

type of clinical presentationdacute coronary syndrome (ACS)<br />

vs. chronic coronary syndrome (CCS). As atherosclerosis is a<br />

systemic disorder, penile circulation might be involved to a<br />

similarly different extent as coronary circulation in ACS vs.<br />

CCS patients. If true, ED prevalence should be low in the<br />

former and high in the latter. 35,36<br />

3. Methods<br />

180 patients with CAD divided into three age-matched groups:<br />

Group 1 (G1, n ¼ 60), ACS with one-vessel disease (1-VD);<br />

Group 2 (G2, n ¼ 60), ACS with 2, 3-VD; Group 3 (G3, n ¼ 60),<br />

chronic stable angina, along with Control group (C, n ¼ 60) was<br />

composed of patients with suspected CAD who were found to<br />

have entirely normal coronary arteries by angiography.<br />

International Index of Erectile Function (IIEF) questionnaires<br />

were used to assess extent of ED. ED as any value 2 months).<br />

We have excluded:<br />

1. Patients with previous percutaneous or surgical myocardial<br />

revascularization procedures.<br />

2. Patients with diseases that could alter sexual activity, such<br />

as liver cirrhosis, renal failure, thyroid disease (hypo- and<br />

hyperthyroidism on replacement treatment), major<br />

depression on long-term pharmacological treatment, and<br />

spinal cord injuries, and those with previous pelvic, penile,<br />

urethral, or prostate trauma or surgery.<br />

3. Patients with primary erectile dysfunction were excluded.<br />

All patients underwent complete routine laboratory tests,<br />

included lipid profile, fasting glucose, and total and freeplasma<br />

testosterone levels. Diagnostic coronary angiography<br />

was carried out in all patients by the standard technique. If<br />

required, percutaneous transluminal coronary angioplasty<br />

(PTCA) or coronary artery bypass graft surgery was carried out<br />

during the hospital stay. Risk factors (when not previously<br />

known) were defined according to the ESC/ACC/AHA guidelines<br />

as follows 38 hypertension as blood pressure >140/<br />

90 mmHg in three consecutive readings, at rest; hypercholesterolemia<br />

as total cholesterol level >200 mg/dL and/or LDL<br />

cholesterol level >130 mg/dL, diabetes as fasting glucose level<br />

>126 mg/dL; obesity as body mass index (BMI) >30 kg/m 2 ; and<br />

family history of CAD as parents with CAD at age


182<br />

indian heart journal 65 (<strong>2013</strong>) 180e186<br />

the indexes obtained for the two legs was used as the measure<br />

of disease severity. 42<br />

The Narayana medical college ethics committee approved<br />

the study protocol and each patient gave written informed<br />

consent.<br />

IIEF-EFD questionnaire for ED (questions 1e5 and 15)<br />

(1) Q: how often were you able to get an erection during sexual<br />

activity? A: no sexual activity (0), almost never/never (1), a<br />

few times (much less than half of the time) (2), sometimes<br />

(about half of the time) (3), most times (much more than<br />

half the time) (4), almost always/always (5).<br />

(2) Q: when you had an erection with sexual stimulation, how<br />

often were your erections hard enough for penetration? A:<br />

no sexual activity (0), almost never/never (1), a few times<br />

(much less than half of the time) (2), sometimes (about half<br />

of the time) (3), most times (much more than half the time)<br />

(4), almost always/always (5).<br />

(3) Q: when you attempted sexual intercourse, how often<br />

were you able to penetrate your partner? A: no sexual activity<br />

(0), almost never/never (1), a few times (2), sometimes<br />

(about half of the time) (3), most times (much more<br />

than half the time) (4), almost always/always (5).<br />

(4) Q: during sexual intercourse, how difficult was it to<br />

maintain your erection after you had penetrate your<br />

partner? A: no sexual activity (0), almost never/never (1), a<br />

few times (2), sometimes (about half of the time) (3), most<br />

times (much more than half the time) (4), almost always/<br />

always (5).<br />

(5) Q: during sexual intercourse, how difficult was it to<br />

maintain your erection to completion of intercourse? A:<br />

did not attempt intercourse (0), extremely difficult (1), very<br />

difficult (2), difficult (3), slightly difficult (4), not difficult (5).<br />

(6) Q: how do you rate your confidence that you could get and<br />

keep an erection? A: very low (1), low (2), moderate (3), high<br />

(4), very high (5)<br />

Fig. 1 e Schematic drawing of the GENSINI score (left). The<br />

method assigns a different severity score depending on the<br />

degree of stenosis, its location (proximal, middle or distal<br />

tract) along the target vessel and the type of coronary<br />

vessel involved (left anterior descending, left CX or RCA).<br />

An example of Gensini score calculation is shown on the<br />

right part of the figure. MLCA, main left coronary artery;<br />

LAD, left anterior descending; CFx, left circumflex; RCA,<br />

right coronary artery.<br />

3.1. Quantitative coronary angiography<br />

3.3. Erectile function evaluation<br />

Coronary angiography analysis was performed by the<br />

cardiologist who is unaware of the patient’s ED. IIEF-EFD<br />

questionnaire, using ARTREK Quantum IC (Image Comm.<br />

System Inc, Sunnyvale, CA, USA). 39 The outer diameter of<br />

the contrast-filled catheter was used for calibration. The lesions<br />

were analyzed in multiple projections, and reference<br />

vessel diameter, minimal lumen diameter, and percent<br />

diameter stenosis were measured from the ‘worst’ angiographic<br />

view. Significant angiographic narrowing was<br />

defined as >50% diameter stenosis involving either one<br />

major epicardial vessel at any site or any collaterals with<br />

>0.3 mm diameter. Patients were classified as having 1-VD,<br />

2-VD, or 3-VD, if they had a single lesion in 1, 2, or 3 coronary<br />

vessels.<br />

3.4. Statistical analysis<br />

3.2. Gensini’s score<br />

The method assigns a different severity score depending on<br />

the degree of stenosis, its location (proximal, middle or distal<br />

tract) along the target vessel and the type of coronary vessel<br />

Erectile function was evaluated by IIEF-EFD, a validated<br />

15-item self-administered questionnaire. 41 Erectile function is<br />

specifically addressed by six questions that form the so called<br />

‘erectile function domain’ of the questionnaire. Each question<br />

is scored 0 to 5. ED is defined as any value


indian heart journal 65 (<strong>2013</strong>) 180e186 183<br />

Table 1 e Baseline characteristics with risk factors.<br />

Control (n ¼ 60) Gr-I (n ¼ 60) Gr-II (n ¼ 60) Gr-III (n ¼ 60) p value<br />

Age (years) 48.5 9 52 8.4 53 8.3 55.4 5.7 0.21<br />

BMI (kg/m 2 ) 26.7 1.2 26.9 1.3 26.4 1.3 26.9 2.1 0.86<br />

Symptom onset (months) 28 12 22 13 18 12 16 9 0.008<br />

Risk factors<br />

Hypertension 57% 56% 54% 55% 0.13<br />

Diabetes 15% 16% 32% 38% 0.06<br />

Hypercholesterolemia 61% 78% 76% 84% 0.06<br />

Smoking 28% 45% 52% 58% 0.08<br />

Obesity 12% 15% 21% 18% 0.07<br />

F/H of CAD 6% 28% 38% 29% 0.005<br />

>3 Risk factor 26% 42% 48% 52% 0.52<br />

mean þ SD, unless otherwise stated. A two tailed P-value<br />

3 risk factors. Overall ED<br />

prevalence was 47%. When separately considered, ED prevalence<br />

was 24%, 56%, and 64% in G1, G2, and G3, respectively<br />

(p < 0.0001 for G1 vs. G2 and G1 vs. G3; p < 0.45 for G2 vs. G3).<br />

ED prevalence in Controls was 22%. ED prevalence was lower<br />

in G1 vs. G3 (24 vs. 64%, p


184<br />

indian heart journal 65 (<strong>2013</strong>) 180e186<br />

70<br />

70<br />

ED prevalence (%)<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

C G1 ACS G2 ACS G3 CCS<br />

60<br />

65<br />

66<br />

62.5<br />

50<br />

52 58<br />

40<br />

30<br />

20 22<br />

10<br />

0<br />

1VD 2VD 3VD 1VD 2VD 3VD<br />

Fig. 2 e Prevalence of ED in the four groups of patients. ACS CSA<br />

Fig. 3 e Prevalence of ED in the ACS and CSA groups.<br />

5. Discussion<br />

ED prevalence (%)<br />

A significant proportion of men with ED exhibit early signs of<br />

CAD, and this group may develop more severe CAD than men<br />

without ED. Prevalence of ED differs across subsets of patients<br />

with CAD and is related to extent of CAD. In group I, ED<br />

prevalence was 24%. This value was similar to that obtained in<br />

age-matched controls with normal coronary arteries. 15 Thus,<br />

most patients with ACS and 1-VD do not complain of ED as<br />

result of an overall low coronary and penile atherosclerotic<br />

burden.<br />

The finding that patients with CCS and 1-VD had higher ED<br />

rate (65 vs. 22%, p < 0.0001) when compared with patients with<br />

ACS and 1-VD, confirms the role of different pathophysiological<br />

background and related atherosclerotic burden at work in<br />

CCS (Fig. 3). Infact, multivariate analysis showed that patients<br />

with CCS presentation had a 2.3-fold increase in relative risk<br />

of ED when compared with those with ACS, independently of<br />

other conventional risk factors. The lower ankle-brachial<br />

index (0.98 þ 0.10 vs.0.80 0.28, p < 0.0001), an accurate and<br />

reliable marker of generalized atherosclerosis, supported a<br />

more advanced vascular involvement in CCS. The time interval<br />

between the onset of ED symptoms and the occurrence<br />

of CAD symptoms and cardiovascular events is estimated at<br />

2e3 years and 3e5 years, respectively; this interval allows for<br />

risk factor reduction.<br />

According to this finding, we evaluated whether ED may<br />

predict coronary artery involvement in ACS. Interestingly<br />

enough, this suggests that the IIEF questionnaire may be a<br />

useful ‘bedside’ test to predict the extension of CAD in ACS:<br />

according to positive predictive value seven out of 10 patients<br />

with ED turned out to have angiographic multivessel disease.<br />

ED-coronary atherosclerosis’ relationship by assessing ED<br />

rate according to CAD extension is being evaluated in this<br />

Table 3 e ED prevalence.<br />

ACS<br />

CCS<br />

1 VD 2 VD 3VD 1VD 2VD 3VD<br />

ED prevalence (%) 22 52 58 65 62.5 66<br />

study. Interesting enough, having 2- or 3-VD did not significantly<br />

increased ED prevalence as compared to 1-VD in both<br />

ACS and CCS patients with similar age (Fig. 2) suggesting ED as<br />

a sort of ‘on-off’ phenomenon that we hypothesized takes<br />

place when 0.50% angiographic obstruction of at least one<br />

major coronary vessel occurs. If true, having 2- or 3-VD would<br />

not add to ED prevalence. Almost 30% of patients with proved<br />

CAD did not complain of ED. Age may be an explanation. We<br />

found age to be independent predictor of ED in the whole<br />

study patient population, with a 10% per patient increase in<br />

the yearly relative risk of ED. ED significantly increased over<br />

time being 30% under 50 years and close to 100% over 60 years<br />

of age. At any age ED rate was similar regardless extent of<br />

CAD, confirming the ‘on-off’ phenomenon.<br />

We found that severe ED (a score


indian heart journal 65 (<strong>2013</strong>) 180e186 185<br />

ED prevalence is related to extent of CAD. (3) ED symptoms<br />

come prior to CAD symptoms in virtually all patients with a<br />

mean time-interval of 3 years. (4) All men with ED should<br />

undergo a thorough medical assessment, including testosterone,<br />

fasting lipids, fasting glucose and blood pressure<br />

measurement. (5) Following assessment, patients should be<br />

stratified according to the risk of future cardiovascular events.<br />

(6) Those at high risk of cardiovascular disease should be<br />

evaluated by stress testing with selective use of computed<br />

tomography (CT) or coronary angiography. (7) Improvement in<br />

cardiovascular risk factors such as weight loss and increased<br />

physical activity has been reported to improve erectile function.<br />

(8) In men with ED, hypertension, diabetes and hyperlipidemia<br />

should be treated aggressively, bearing in mind the<br />

potential side effects. (9) Management of ED is secondary to<br />

stabilizing cardiovascular function, and controlling cardiovascular<br />

symptoms and exercise tolerance should be established<br />

prior to initiation of ED therapy. (10) Clinical evidence<br />

supports the use of phosphodiesterase 5 (PDE5) inhibitors as<br />

first-line therapy in men with CAD and comorbid ED and those<br />

with diabetes and ED. (11) Review of cardiovascular status and<br />

response to ED therapy should be performed at regular<br />

intervals.<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Lue TF. Erectile dysfunction. N Engl J Med. 2000;342:1802e1813.<br />

2. Aytac IA, McKinlay JB, Krane RJ. The likely worldwide increase<br />

in erectile dysfunction between 1995 and 2025 and some<br />

possible policy consequences. Br J Urol Int. 1999;84:50e56.<br />

3. Feldman HA, Goldstein I, Hatzichristou D, Krane RJ,<br />

McKinlay JB. Impotence and its medical and psychological<br />

correlates: results of the Massachusset Male Aging Study.<br />

J Urol. 1994;151:54e61.<br />

4. Rosen RC, Cappelleri JC, Smith MD, Lipsky J, Pena BM.<br />

Development and evaluation of an abridged, 5-item version of<br />

the International Index of Erectile Function (IIEF-5) as a<br />

diagnostic tool for erectile dysfunction. Int J Impot Res.<br />

1999;11:319e326.<br />

5. Chiurlia E, D’Amico R, Ratti C, Granata AR, Romagnoli R,<br />

odena MG. Subclinical coronary artery atherosclerosis in<br />

patients with erectile dysfunction. J Am Coll Cardiol.<br />

2005;46:1503e1506.<br />

6. Vlachopoulos C, Rokkas K, Ioakeimidis N, Stefanadis C.<br />

Inflammation, metabolic syndrome, erectile dysfunction, and<br />

coronary artery disease: common links. Eur Urol.<br />

2007;52:1590e1600.<br />

7. Solomon H, Man JW, Wierzbicki AS, Jackson G. Relation of<br />

erectile dysfunction to angiographic artery disease. Am J<br />

Cardiol. 2002;91:230e231.<br />

8. Montorsi F, Briganti A, Salonia A, et al. Erectile dysfunction<br />

prevalence, time of onset and association with risk factors in<br />

300 consecutive patients with acute chest pain and<br />

angiographically documented coronary artery disease. Eur<br />

Urol. 2003;44:360e365.<br />

9. Kloner RA, Mullin S, Shook T, et al. Erectile dysfunction in the<br />

cardiac patient: how common and should we treat? J Urol.<br />

2003;170:S46eS50.<br />

10. Wabrek AJ, Burchell C. Male sexual dysfunction associated<br />

with coronary artery disease. Arch Sex Behav. 1980;9:69e75.<br />

11. Diokno AC, Brown MB, Herzog R. Sexual function in the<br />

elderly. Arch Intern Med. 1990;150:197e200.<br />

12. Dhabuwala CB, Kumar A, Pierce JM. Myocardial infarction and<br />

its influence on male sexual function. Arch Sex Behav.<br />

1986;15:499e504.<br />

13. Welt FGP, Simon DI. Atherosclerosis and claque rupture.<br />

Catheter Cardiovasc Interv. 2001;53:56e63.<br />

14. Fedele D, Bortolotti A, Coscelli C, et al, on behalf of Gruppo<br />

Italiano Studio Deficit Erettile nei Diabetici. Erectile<br />

dysfunction in Type 1 and Type 2 diabetics in Italy. Int J<br />

Epidemiol. 2000;29:524e531.<br />

15. Feldman JA, Goldstein I, Hatzichristou DG, et al. Impotence<br />

and its medical and physiological correlates: results of the<br />

Massachusetts Male Aging Study. J Urol. 1994;151:54e61.<br />

16. Adams PF, <strong>Mar</strong>ano MA. Current estimates from the National<br />

Health Interview Survey. Vital Health Stat. 1995;10:83e84.<br />

17. Greenstein A, Chen J, Miller H, et al. Does severity of ischemic<br />

coronary disease correlate with erectile function? Int J Impot<br />

Res. 1997;9:123e126.<br />

18. Koistinen MJ. Prevalence of asymptomatic myocardial<br />

ischaemia in diabetic subjects. BMJ. 1990;301:92e95.<br />

19. Ashley EA, Raxval V, Finlay M, et al. Diagnosing coronary<br />

artery disease in diabetic patients. Diabetes Metab Res Rev.<br />

2002;18:201e208.<br />

20. Billups KL, Kaiser DR, Kelly AS, et al. Relation of C-reactive<br />

protein and other cardiovascular risk factors to penile<br />

vascular disease in men with erectile dysfunction. Int J Impot<br />

Res. 2003;15:231e236.<br />

21. Roumeguere T, Wespes E, Carpentier Y, Hoffmann P,<br />

Schulman CC. Erectile dysfunction is associated with a high<br />

prevalence of hyperlipidemia and coronary heart disease risk.<br />

Eur Urol. 2003;44:355e359.<br />

22. El-Sakka AI, Morsy AM, Fagih BI, Nassar AH. Coronary artery<br />

risk factors in patients with erectile dysfunction. J Urol.<br />

2004;172:251e254.<br />

23. Vlachopoulos C, Aznaouridis K, Ioakeimidis N, et al.<br />

Unfavourable endothelial and inflammatory state in erectile<br />

dysfunction patients with or without coronary artery disease.<br />

Eur <strong>Heart</strong> J. 2006;27:2640e2648. 1503e6.<br />

24. Min JK, Williams KA, Okwuosa TM, Bell GW, Panutich MS,<br />

Ward RP. Prediction of coronary heart disease by erectile<br />

dysfunction in men referred for nuclear stress testing. Arch<br />

Intern Med. 2006;166:201e206.<br />

25. Montorsi P, Ravagnani PM, Galli S, et al. Association between<br />

erectile dysfunction and coronary artery disease. Role of<br />

coronary clinical presentation and extent of coronary<br />

vessels involvement: the COBRA trial. Eur <strong>Heart</strong> J. 2006;27:<br />

2632e2639.<br />

26. Ward RP, Weiner J, Taillon LA, Ghani SN, Min JK, Williams KA.<br />

Comparison of findings on stress myocardial perfusion<br />

imaging in men with versus without erectile dysfunction and<br />

without prior heart disease. Am J Cardiol. 2008;101:502e505.<br />

27. Salem S, Abdi S, Mehrsai A, et al. Erectile dysfunction severity<br />

as a risk predictor for coronary artery disease. J Sex Med.<br />

2009;6:3425e3432.<br />

28. Montorsi P, Ravagnani PM, Galli S, et al. Association between<br />

erectile dysfunction and coronary artery disease: matching<br />

the right target with the right test in the right patient. Eur<br />

Urol. 2006;50:721e731.<br />

29. Baumhakel M, Bohm M. Erectile dysfunction correlates with<br />

left ventricular function and precedes cardiovascular events<br />

in cardiovascular high-risk patients. Int J Clin Pract. 2007;61:<br />

361e366.


186<br />

indian heart journal 65 (<strong>2013</strong>) 180e186<br />

30. Hodges LD, Kirby M, Solanki J, O’Donnell J, Brodie DA. The<br />

temporal relationship between erectile dysfunction and<br />

cardiovascular disease. Int J Clin Pract. 2007;61:2019e2025.<br />

31. Chew KK, Finn J, Stuckey B, et al. Erectile dysfunction as a<br />

predictor for subsequent atherosclerotic cardiovascular<br />

events: findings from a linked-data study. J Sex Med.<br />

2010;7:192e202.<br />

32. Ponholzer A, Temml C, Obermayr R, Wehrberger C,<br />

Madersbacher S. Is erectile dysfunction an indicator for<br />

increased risk of coronary heart disease and stroke? Eur Urol.<br />

2005;48:512e518.<br />

33. Thompson IM, Tangen CM, Goodman PJ, Probstfield JL,<br />

Moinpour CM, Coltman CA. Erectile dysfunction and<br />

subsequent cardiovascular disease. JAMA.<br />

2005;294:2996e3002.<br />

34. Schounten BW, Bohnen AM, Bosch JL, et al. Erectile<br />

dysfunction prospectively associated with cardiovascular<br />

disease in the Dutch general population: results from the<br />

Krimpen Study. Int J Impot Res. 2008;20:92e99.<br />

35. Montorsi P, Montorsi F, Schulman C. Is erectile dysfunction<br />

the tip of the iceberg of a systemic vascular disorder? Eur Urol.<br />

2003;44:352e354.<br />

36. Montorsi P, Ravagnani P, Galli S, et al. Association between<br />

erectile dysfunction and coronary artery disease: a case<br />

report study. J Sex Med. 2005;2:575e582.<br />

37. De Backer G, Ambrosioni E, Borch-Johnsen K, et al. European<br />

guidelines on cardiovascular disease prevention in clinical<br />

practice. Eur <strong>Heart</strong> J. 2003;24:1601e1610.<br />

38. Braunwald E. Unstable angina. Circulation. 1989;80:410e414.<br />

39. Mancini GBJ, Simon SB, McGillem MJ, LeFree MT, Friedman HZ,<br />

Vogel RA. Automated quantitative coronary arteriography:<br />

morphologic and physiologic validation in vivo of a rapid<br />

digital angiographic method. Circulation. 1987;75:452e460.<br />

40. Gensini G. A more meaningful scoring system for<br />

determining the severity of coronary artery disease. Am J<br />

Cardiol. 1983;51:606.<br />

41. Jackson G, Betteridge J, Dean J, et al. A systematic approach to<br />

erectile dysfunction in the cardiovascular patient: a<br />

consensus statement e update 2002. Int J Clin Pract.<br />

2002;56:663e671.<br />

42. Bird CE, Criqui MH, Fronek A, Denenberg JO, Klauber MR,<br />

Langer RD. Quantitative and qualitative progression of<br />

peripheral arterial disease by non-invasive testing. Vasc Med.<br />

1999;4:15e21.


indian heart journal 65 (<strong>2013</strong>) 219e228 221<br />

(2.15 0.27 mm vs. 2.25 0.24 mm; p ¼ 0.003). The majority<br />

(89%) of lesions involved vessels with a diameter


222<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

patients without baseline “SCD-HeFT criteria” (left ventricular<br />

ejection fraction >0.35 or NYHA class I), 125 were<br />

evaluated after 5.5 2 months. Of these 227 patients, 13<br />

(10%) developed “SCD-HeFT criteria” (group B1), 111 (89%)<br />

remained without “SCD-HeFT criteria” (group B2), and 1<br />

(1%) had worsened to NYHA class IV. The 10-year mortality/heart<br />

transplantation and sudden death/sustained<br />

ventricular arrhythmia rate was 57% and 37% in group A1,<br />

23% and 20% in group A2 (p < 0.001 for mortality/heart<br />

transplantation and p e 0.014 for sudden death/sustained<br />

ventricular arrhythmia vs. group A1), 45% and 41% in group<br />

B1 ( p e NS vs. group A1), 16% and 14% in group B2 ( p e NS<br />

vs. group A2), respectively.<br />

Conclusion: Two thirds of patients with idiopathic<br />

dilated cardiomyopathy and “SCD-HeFT criteria” at presentation<br />

did not maintain implantable cardioverterdefibrillator<br />

indications 3e9 months later with optimal<br />

medical therapy. Their long-term outcome was excellent,<br />

similar to that observed for patients who had never met the<br />

“SCD-HeFT criteria.”<br />

1. Perspective<br />

Since the publication of the SCD-HeFT and the DEFINITE<br />

trials, treatment with ICD for the primary prevention of<br />

sudden cardiac death (SCD) has been extended to patients<br />

with idiopathic dilated cardiomyopathy (IDC), who have a<br />

LVEF of 0.35 and who are classified as NYHA II or III (“SCD-<br />

HeFT criteria,” class I B indication). The appropriate timing<br />

for ICD implantation, however, is still uncertain. Current<br />

guidelines suggest that an ICD should be considered in<br />

addition to medical therapy, but many patients are treated<br />

with an ICD without evidence-based indications, mainly<br />

because of newly diagnosed heart failure and before treatment<br />

optimization. This study evaluated the proportion of<br />

patients with and without potential indications for ICD<br />

implantation at presentation and the long-term prognosis of<br />

patients with initial ICD indications but who improved after<br />

optimization of medical treatment. It also compared the<br />

long-term outcome of “improved” patients to those maintaining<br />

“SCD-HeFT criteria” and those who never met “SCD-<br />

HeFT criteria.”<br />

This trial included only patients who were not on beta<br />

blockers. After initial assessment, optimization of medical<br />

treatment was achieved with gradually up-titrating doses of<br />

beta blockers and ACEI/ARB at the highest tolerated dose over<br />

a period of 3e9 months.<br />

The main results of the present study are: 1) 50% patients<br />

had SCD-HeFT criteria at first assessment and would have<br />

otherwise received an ICD. 2) 2/3rd of patients with SCD-HeFT<br />

criteria at baseline “improved” and no longer maintained SCD-<br />

HeFT criteria 5.5 months after starting beta blocker and ACEI<br />

treatment. 3) The long-term SCD were similar in “improved”<br />

patients and in those without SCD-HeFT criteria, suggesting<br />

ICD implantation should not be done in most patients with<br />

low LVEF and HF symptoms before optimization of medical<br />

treatment. 4) SCD (4 patients e 2%) was similar in patients<br />

both with and without SCD-HeFT criteria before second<br />

evaluation at 3e9 months, confirming the difficulty of stratifying<br />

risk of SCD at first evaluation.<br />

This study emphasizes how important is the optimization<br />

of medical therapy in patients initially presenting with ICD<br />

indications and ICD implantation can be avoided in the<br />

majority of such patients. Even in USA, nearly 22.5% of<br />

patients with an ICD did not meet the evidence-based criteria<br />

for implantation, mainly because of newly diagnosed<br />

HF (62%). Such unnecessary ICD implantations should be<br />

avoided because of economic issues (especially in a developing<br />

country like India), the risk of complications associated<br />

with implantation and inappropriate shocks (in w25%<br />

of patients).<br />

What then is the waiting period for ICD implantation after<br />

onset of HF symptoms in patients with LVEF 35%? Well, we<br />

have no clear-cut answer. Data from DEFINITE trial suggest<br />

early ICD implantation (


indian heart journal 65 (<strong>2013</strong>) 234e235<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Letter to the Editor<br />

Association between blood glucose level and in-hospital<br />

mortality in patients with acute myocardial infarction<br />

Dear Editor, Diabetes mellitus increases cardiovascularrelated<br />

morbidity and mortality and high blood glucose even<br />

before it reaches the threshold to diagnose diabetes is associated<br />

with higher cardiovascular risk.<br />

Several studies previously reported that adverse cardiac<br />

events increased in patients with acute myocardial infarction.<br />

However, the association between admission blood glucose<br />

level and cardiac-related mortality and morbidity was not<br />

studied in Iranian population.<br />

Therefore, we conducted a study to assess the association<br />

between on admission blood glucose level and in-hospital<br />

cardiac mortality and prognosis in these patients.<br />

The study designed prospectively and performed on<br />

patients presenting to the emergency department of our<br />

center with acute ST elevation myocardial infarction<br />

(STEMI) between <strong>Apr</strong>il, 2008 and <strong>Apr</strong>il, 2010. STEMI was<br />

diagnosed based on the acute rise and gradual fall of cardiac<br />

enzymes with ischemic symptoms and ECG changes (ST<br />

segment elevation or new left bundle branch block). Exclusion<br />

criteria were severe co-morbidities, history of valvular<br />

heart disease, and low ejection fraction before infarction<br />

(EF


indian heart journal 65 (<strong>2013</strong>) 234e235 235<br />

Hooman Bakhshandeh, Tahereh Zandi, Majid Maleki,<br />

Anoushiravan Vakili-Zarch<br />

Rajaei <strong>Heart</strong> Center, Tehran University of Medical Sciences, Iran<br />

*Corresponding author. Assistant Professor of Cardiology,<br />

Valiasr Ave., Niyayesh Exp., Shaheed Rajaei Cardiovascular<br />

and Medical Research Center, Tehran, Iran.<br />

Tel.: þ98 (21) 23922176; fax: þ98 (21) 22055594.<br />

E-mail address: negar70049@gmail.com (N. Salehi)<br />

Available online 26 February <strong>2013</strong><br />

0019-4832/$ e see front matter<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights<br />

reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.015


indian heart journal 65 (<strong>2013</strong>) 234e235<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Letter to the Editor<br />

Association between blood glucose level and in-hospital<br />

mortality in patients with acute myocardial infarction<br />

Dear Editor, Diabetes mellitus increases cardiovascularrelated<br />

morbidity and mortality and high blood glucose even<br />

before it reaches the threshold to diagnose diabetes is associated<br />

with higher cardiovascular risk.<br />

Several studies previously reported that adverse cardiac<br />

events increased in patients with acute myocardial infarction.<br />

However, the association between admission blood glucose<br />

level and cardiac-related mortality and morbidity was not<br />

studied in Iranian population.<br />

Therefore, we conducted a study to assess the association<br />

between on admission blood glucose level and in-hospital<br />

cardiac mortality and prognosis in these patients.<br />

The study designed prospectively and performed on<br />

patients presenting to the emergency department of our<br />

center with acute ST elevation myocardial infarction<br />

(STEMI) between <strong>Apr</strong>il, 2008 and <strong>Apr</strong>il, 2010. STEMI was<br />

diagnosed based on the acute rise and gradual fall of cardiac<br />

enzymes with ischemic symptoms and ECG changes (ST<br />

segment elevation or new left bundle branch block). Exclusion<br />

criteria were severe co-morbidities, history of valvular<br />

heart disease, and low ejection fraction before infarction<br />

(EF


indian heart journal 65 (<strong>2013</strong>) 234e235 235<br />

Hooman Bakhshandeh, Tahereh Zandi, Majid Maleki,<br />

Anoushiravan Vakili-Zarch<br />

Rajaei <strong>Heart</strong> Center, Tehran University of Medical Sciences, Iran<br />

*Corresponding author. Assistant Professor of Cardiology,<br />

Valiasr Ave., Niyayesh Exp., Shaheed Rajaei Cardiovascular<br />

and Medical Research Center, Tehran, Iran.<br />

Tel.: þ98 (21) 23922176; fax: þ98 (21) 22055594.<br />

E-mail address: negar70049@gmail.com (N. Salehi)<br />

Available online 26 February <strong>2013</strong><br />

0019-4832/$ e see front matter<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights<br />

reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.02.015


indian heart journal 65 (<strong>2013</strong>) 219e228<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

<strong>Journal</strong> Reviews<br />

Gregg W. Stone, Akiko Maehara, Bernhard Witzenbichler,<br />

Jacek Godlewski, Helen Parise, Jan-Henk E. Dambrink,<br />

Andrzej Ochala, Trevor W. Carlton, Ecaterina Cristea,<br />

Steven D. Wolff, Sorin J. Brener, Saqib Chowdhary,<br />

Magdi El-Omar, Thomas Neunteufl, D. Christopher Metzger,<br />

Theodore Karwoski, Jose M. Dizon, Roxana Mehran, C.<br />

Michael Gibson, for the INFUSE-AMI Investigators, Intracoronary<br />

abciximab and aspiration thrombectomy in patients<br />

with large anterior myocardial infarction: the INFUSE-AMI<br />

randomized trial. JAMA 307 (17) (2012) 1817e1826<br />

1. Context<br />

Thrombus embolization during percutaneous coronary intervention<br />

(PCI) in ST-segment elevation myocardial infarction<br />

(STEMI) is common and results in suboptimal myocardial<br />

perfusion and increased infarct size. Two strategies proposed<br />

to reduce distal embolization and improve outcomes after<br />

primary PCI are bolus intracoronary abciximab and manual<br />

aspiration thrombectomy.<br />

2. Objective<br />

To determine whether bolus intracoronary abciximab, manual<br />

aspiration thrombectomy, or both reduce infarct size in<br />

high-risk patients with STEMI.<br />

3. Design, setting, and patients<br />

Between November 28, 2009, and December 2, 2011, 452<br />

patients presenting at 37 sites in 6 countries within 4 h of<br />

STEMI due to proximal or mid left anterior descending artery<br />

occlusion undergoing primary PCI with bivalirudin anticoagulation<br />

were randomized in an open-label, 22 factorial<br />

design to bolus intracoronary abciximab delivered locally at<br />

the infarct lesion site vs no abciximab and to manual aspiration<br />

thrombectomy vs no thrombectomy.<br />

4. Interventions<br />

A 0.25 mg/kg bolus of abciximab was administered at the site<br />

of the infarct lesion via a local drug delivery catheter. Manual<br />

aspiration thrombectomy was performed with a 6 F aspiration<br />

catheter.<br />

5. Main outcome measures<br />

Primary end point: infarct size (percentage of total left ventricular<br />

mass) at 30 days assessed by cardiac magnetic<br />

resonance imaging (cMRI) in the abciximab vs no abciximab<br />

groups (pooled across the aspiration randomization); major<br />

secondary end point: 30-day infarct size in the aspiration vs<br />

no aspiration groups (pooled across the abciximab<br />

randomization).<br />

6. Results<br />

Evaluable cMRI results at 30 days were present in 181 and 172<br />

patients randomized to intracoronary abciximab vs no<br />

abciximab, respectively, and in 174 and 179 patients<br />

randomized to manual aspiration vs. no aspiration, respectively.<br />

Patients randomized to intracoronary abciximab<br />

compared with no abciximab had a significant reduction in<br />

30-day infarct size (median, 15.1%; interquartile range [IQR],<br />

6.8%e22.7%; n ¼ 181, vs. 17.9% [IQR, 10.3%e25.4%]; n ¼ 172;<br />

p ¼ 0.03). Patients randomized to intracoronary abciximab<br />

also had a significant reduction in absolute infarct mass<br />

(median, 18.7 g [IQR, 7.4e31.3 g]; n ¼ 184, vs. 24.0 g [IQR,<br />

12.1e34.2 g]; n ¼ 175; p ¼ 0.03) but not abnormal wall motion<br />

score (median, 7.0 [IQR, 2.0e10.0]; n ¼ 188, vs. 8.0 [IQR,<br />

3.0e10.0]; n ¼ 184; p ¼ 0.08). Patients randomized to aspiration<br />

thrombectomy vs no aspiration had no significant difference<br />

in infarct size at 30 days (median, 17.0% [IQR, 9.0%e22.8%];<br />

n ¼ 174, vs. 17.3% [IQR, 7.1%e25.5%]; n ¼ 179; p ¼ 0.51), absolute<br />

infarct mass (median, 20.3 g [IQR, 9.7e31.7 g]; n ¼ 178, vs. 21.0 g<br />

[IQR, 9.1e34.1 g]; n ¼ 181; p ¼ 0.36), or abnormal wall motion<br />

score (median, 7.5 [IQR, 2.0e10.0]; n ¼ 186, vs. 7.5 [IQR,<br />

2.0e10.0]; n ¼ 186; p ¼ 0.89).<br />

7. Conclusion<br />

In patients with large anterior STEMI presenting early after<br />

symptom onset and undergoing primary PCI with bivalirudin<br />

anticoagulation, infarct size at 30 days was significantly<br />

reduced by bolus intracoronary abciximab delivered to<br />

the infarct lesion site but not by manual aspiration<br />

thrombectomy.<br />

8. Perspective<br />

In this multicenter, prospective, randomized trial in patients<br />

with large anterior STEMI presenting early after infarct onset<br />

and undergoing primary PCI with bivalirudin anticoagulation,<br />

the principal findings were: 1) bolus intracoronary abciximab<br />

delivered to the site of the lesion via a clearway catheter significantly<br />

but modestly reduced the infarct size at 30 days 2)<br />

thrombus aspiration with export catheter had no effect on<br />

infarct size and 3) indices of myocardial reperfusion, ST


220<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

resolution (STR) and 30-day MACE (w7% in all groups)/stent<br />

thrombosis/bleeding were not significantly different between<br />

the randomized groups.<br />

Two of the strongest baseline determinants of infarct size<br />

are: 1) anterior MI location and 2) abnormal TIMI flow. This<br />

trial was limited to patients with proximal or mid LAD<br />

occlusion and TIMI 0e2 flow. Moreover, it only enrolled<br />

patients who could be treated early, in whom time window for<br />

effective myocardial salvage had not closed. The median time<br />

from symptom onset to hospital arrival was only 99 min and<br />

the median D-to-B time was 45 min. Thus the study population<br />

represents a highly selected cohort of patients with<br />

large anterior MI, in whom infarct size reduction should be<br />

feasible given early presentation and rapid treatment. Infarct<br />

size was assessed by cMRI, which strongly correlates with<br />

subsequent mortality. Unlike prior studies, which measured<br />

infarct size at 2e7 days (a period during which substantial<br />

myocardial edema is present, thereby interfering with<br />

assessment of viable myocardium) in this study cMRI was<br />

done at 30 days when much of myocardial edema had<br />

resolved.<br />

These results need to be placed in the context of previous<br />

studies. A meta-analysis of 6 RCT’s (1246 patients) reported<br />

enhanced survival with bolus intracoronary abciximab.<br />

However, the recent AIDA-STEMI trial (2065 patients) found<br />

nearly identical rates of MACE with bolus intracoronary and<br />

intravenous abciximab. However, this trial differs from these<br />

earlier studies in many ways: 1) unlike prior studies which<br />

included routine post-PCI intravenous abciximab infusion in<br />

both the groups, in this trial only bolus intracoronary abciximab<br />

was given in the randomized groups. 2) In all prior trials<br />

(including AIDA-STEMI), intracoronary abciximab was infused<br />

proximally through the guide catheter thereby limiting its<br />

penetration into occlusive thrombus and allowing spillage of<br />

the drug to LCx or backflow into the aorta. In contrast, the<br />

local drug delivery catheter (clearway catheter) used in this<br />

study achieves high intra-clot concentration of abciximab at<br />

the site of LAD occlusion and prolongs drug residence time,<br />

which may enhance platelet disaggregation and thrombus<br />

resolution. In the present study, an abciximab bolus delivered<br />

directly to the infarct lesion site (without a 12-hour infusion)<br />

reduced infarct size at 30 days in patients with anterior STEMI<br />

reperfused early.<br />

Regarding aspiration thrombectomy, in TAPAS, 1071<br />

patients with anterior and non-anterior STEMI who presented<br />

within 12 h of symptoms at a single-center were randomized<br />

to manual aspiration vs. no aspiration before primary PCI;<br />

aspiration resulted in modest improvements in MBG and STR<br />

but a marked reduction in 1-year mortality. Other trials have<br />

reported conflicting results, and in contrast to single-center<br />

studies, multicenter aspiration trials have been largely negative.<br />

Moreover, in TAPAS, aspiration did not reduce infarct<br />

size as measured by cardiac biomarkers, calling into question<br />

the mechanism underlying the survival benefit. The present<br />

multicenter trial, in which only patients presenting early with<br />

anterior MI and coronary anatomy optimal for aspiration were<br />

enrolled, and in which cMRI was used to assess infarct size at<br />

30 days was specifically designed to overcome many of the<br />

limitations from these earlier studies. The fact that manual<br />

thrombus aspiration did not reduce infarct size in this study<br />

makes a substantial clinical benefit unlikely, questioning its<br />

routine use in STEMI.<br />

9. Our opinion<br />

Regarding use of GPIIb/IIIa inhibitors: a) I/V bolus and infusion<br />

is to be discouraged because it achieves very little intra-clot<br />

concentration and also increases the risk of systemic bleeding.<br />

b) Only bolus intracoronary drug should be used, that too<br />

not into the guide catheter, but via a clearway catheter (we can<br />

use a simple PTCA balloon by making multiple holes on its<br />

surface, in case clearway catheter is not available).<br />

Regarding manual aspiration via Export catheter: a) the<br />

symptom onset to hospital arrival and the D-to-B time were<br />

substantially shorter in this study which is next to impossible<br />

in our context. b) As time passes by after STEMI thrombus<br />

tends to get organized and hence thrombus aspiration might<br />

have some role to play in late presenters of STEMI. However,<br />

the last word in this matter is yet to be written.<br />

Suraj Khanal*<br />

Assistant Professor of Cardiology, Department of Cardiology,<br />

3rd Floor, Block-C, Advanced Cardiac Center, PGIMER,<br />

Chandigarh 160012, India<br />

Ajay Bahl<br />

Associate Professor of Cardiology, PGIMER, Chandigarh, India<br />

*Corresponding author. Tel.: þ91 09878222526.<br />

E-mail address: khanal.s@rediffmail.com<br />

Azeem Latib, Antonio Colombo, Fausto Castriota,<br />

Antonio Micari, Alberto Cremonesi, Francesco De Felice,<br />

Alfredo <strong>Mar</strong>chese, Maurizio Tespili, Patrizia Presbitero,<br />

Gregory A. Sgueglia, Francesca Buffoli, Corrado Tamburino,<br />

Ferdinando Varbella, Alberto Menozzi, A randomized multicentre<br />

study comparing a paclitaxel drug-eluting balloon with<br />

a paclitaxel-eluting stent in small coronary vessels: The<br />

BELLO (Balloon Elution and Late Loss Optimization) study. J<br />

Am Coll Cardiol. 60 (2012) 2473e2480<br />

Objectives: The aim of this study was to evaluate the efficacy<br />

of drug-eluting balloons (DEB) compared with paclitaxel<br />

eluting stents (PES) for the reduction of restenosis in small<br />

vessels.<br />

Background: DEB have been shown to be effective in the<br />

treatment of coronary in-stent restenosis, but data are limited<br />

regarding their efficacy in de-novo disease.<br />

Methods: BELLO (Balloon Elution and Late Loss Optimization)<br />

is a prospective, multicentre trial that randomized 182 patients<br />

with lesions located in small vessels (reference diameter<br />


indian heart journal 65 (<strong>2013</strong>) 194e197 197<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Greenspan M, Iskandrian AS, Segal BL, Kimbiris D, Bemis CE.<br />

Complete occlusion of the left main coronary artery. Am <strong>Heart</strong><br />

J. 1979;98:83e86.<br />

2. Zimmern SH, Rogers WJ, Bream PR, et al. Total occlusion of the<br />

left main coronary artery: the coronary artery surgery study<br />

(CASS) experience. Am J Cardiol. 1982;49:2003e2010.<br />

3. Takagi H, Kawai N, Umemoto T. Stenting versus coronary<br />

artery bypass grafting for unprotected left main coronary<br />

artery disease: a meta-analysis of comparative studies. J Thorac<br />

Cardiovasc Surg. 2009;137:54e57.<br />

4. Morice MC, Serruys PW, Kappetein AP, et al. Outcomes in patients<br />

with de novo left main disease treated with either percutaneous<br />

coronary intervention using paclitaxel-eluting stents or coronary<br />

artery bypass graft treatment in the Synergy between<br />

Percutaneous Coronary Intervention with TAXUS and Cardiac<br />

Surgery (SYNTAX) trial. Circulation. 2010;121:2645e2653.<br />

5. Koster NK, White M. Chronic effort-induced angina as<br />

presentation of a totally occluded left main coronary artery: a<br />

case report and review. Angiology. 2009;60:382e384.<br />

6. Secco GG, <strong>Mar</strong>ino PN, Venegoni L, De Luca G. Percutaneous<br />

revascularization of chronic total occlusion of the left main<br />

coronary artery. Rev Esp Cardiol. 2011;64:431e433.<br />

Book Review<br />

Pediatric Cardiac Intensive Care. Pre and Postoperative<br />

Guidelines, Manoj Luthra. Elsevier A Division of Reed Elsevier<br />

India Pvt Ltd, 3, Tolstoy <strong>Mar</strong>g, New Delhi, India, (2012).<br />

Pages: 314, Price: INR 475/-<br />

Pediatric Cardiac Intensive Care has seen a tremendous<br />

progress over the last few decades in our country. The centers<br />

providing this specialized care have increased steadily with<br />

newer centers being added every year across non-metro cities<br />

too. This has created a requirement of good pediatric cardiac<br />

intensive care services which most often is delivered by the<br />

team of pediatric cardiac surgeon, pediatric cardiac anesthetist,<br />

pediatric cardiologist, intensivist, pediatrician, registrars<br />

and nurses at various levels. With this scenario, there is an<br />

urgent need for a simple yet informative and concise handbook<br />

to enable provision of quality care to some of the sickest<br />

of children with cardiac problems across their peri-operative<br />

and post-operative period.<br />

‘The Manual of Pediatric cardiac intensive care e Pre and<br />

postoperative guidelines’ by Dr Manoj Luthra, an eminent<br />

pediatric cardiac surgeon of our country, is an excellent<br />

handbook which should enable the team of pediatric cardiac<br />

intensive care providers to handle most of the situations<br />

arising in the unit. It has chapters on almost all the relevant<br />

emergencies such as congestive cardiac failure, arrhythmias,<br />

hypertension, pulmonary hypertension, post-operative respiratory<br />

complications, ARDS, ventilator associated pneumonia,<br />

sepsis, seizures, acute kidney injury and<br />

coagulopathies. Besides these emergencies, many basic<br />

physiology topics such as fluid and electrolytes, ABG analysis,<br />

hemodynamic monitoring, parenteral and enteral nutrition<br />

have been discussed very succinctly .It also contains valuable<br />

chapters on important drugs used in the PCCU and has a well<br />

compiled set of appendices at the end. The book is handy, is<br />

well illustrated and the author has kept the language very<br />

simple so that it can be used by the different levels of child<br />

care providers. The book is likely to be useful to anyone<br />

involved in looking after the sick child in the pediatric cardiac<br />

care unit.<br />

It is a difficult task to convert volumes of information<br />

available on pediatric cardiac intensive care to a few hundred<br />

pages. However, the author has created an excellent handbook<br />

on the subject and reflects decades of experience in<br />

practical day to day management in the PCCU and toward<br />

which the book shall go a long way in fulfilling the requirements<br />

of the pediatric cardiac care provider.<br />

B.M. John*, Amit Devgan<br />

Associate Professor, Department of Pediatrics, AFMC,<br />

Sholapur Road, Pune 411040, India<br />

*Corresponding author. Tel.: þ91 09372326660.<br />

E-mail address: drbmj1972@yahoo.com (B.M. John)


228<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

H.M. <strong>Mar</strong>dikar*<br />

Director, Spandan <strong>Heart</strong> Institute & Research Center,<br />

31, Off Chitale <strong>Mar</strong>g, Dhantoli,<br />

Nagpur 440010, India<br />

*Corresponding author. Tel.: þ91 9823082609 (mobile), þ91 (0)<br />

712 2443333; fax: þ91 (0) 712 2443426.<br />

E-mail address: drmardikar@cadindia.co.in<br />

Conclusions: Control patients who crossed over to renal<br />

denervation with the Symplicity system had a significant drop<br />

in blood pressure similar to that observed in patients receiving<br />

immediate denervation. Renal denervation provides safe and<br />

sustained reduction of blood pressure to 1 year.<br />

Clinical trial registration: URL: http://www.clinicaltrials.<br />

gov. Unique identifier: http://www.clinicaltrials.gov. (Circulation.<br />

2012;126:2976e2982.)<br />

Murray D. Esler, Henry Krum, <strong>Mar</strong>kus Schlaich, Roland E.<br />

Schmieder, Michael Böhm, Paul A. Sobotka, for the Symplicity<br />

HTN-2 Investigators. Renal sympathetic denervation for<br />

treatment of drug-resistant hypertension one-year results<br />

from the Symplicity HTN-2 randomized, controlled trial.<br />

Background: Renal sympathetic nerve activation contributes<br />

to the pathogenesis of hypertension. Symplicity HTN-2, a<br />

multicenter, randomized trial, demonstrated that catheterbased<br />

renal denervation produced significant blood pressure<br />

lowering in treatment-resistant patients at 6 months after the<br />

procedure compared with control, medication-only patients.<br />

Longer-term follow-up, including 6-month crossover results,<br />

is now presented.<br />

Methods and results: Eligible patients were on 3 antihypertensive<br />

drugs and had a baseline systolic blood pressure<br />

160 mm Hg (150 mm Hg for type 2 diabetics). After the<br />

6-month primary end point was met, renal denervation in<br />

control patients was permitted. One-year results on patients<br />

randomized to immediate renal denervation (n ¼ 47) and<br />

6-month postprocedure results for crossover patients are<br />

presented. At 12 months after the procedure, the mean fall in<br />

office systolic blood pressure in the initial renal denervation<br />

group ( 28.1 mm Hg; 95% confidence interval, 35.4 to 20.7;<br />

p < 0.001) was similar to the 6-month fall ( 31.7 mm Hg; 95%<br />

confidence interval, 38.3 to 25.0; p < 0.16 versus 6-month<br />

change). The mean systolic blood pressure of the crossover<br />

group 6 months after the procedure was significantly lowered<br />

(from 190.0 19.6 to 166.3 24.7 mm Hg; change, 23.7 27.5;<br />

p < 0.001). In the crossover group, there was 1 renal artery<br />

dissection during guide catheter insertion, before denervation,<br />

corrected by renal artery stenting, and 1 hypotensive<br />

episode, which resolved with medication adjustment.<br />

1. Clinical perspective<br />

The efficacy and safety of the Renal sympathetic Denervation<br />

(RDN) by utilizing the Symplicity Renal Denervation<br />

System in patients with uncontrolled resistant hypertension<br />

have been documented earlier. It has been shown that<br />

activation of the sympathetic nervous system is involved in<br />

the pathogenesis and maintenance of hypertension. Renal<br />

denervation with the Symplicity catheter is a minimally<br />

invasive procedure based on the premise that interruption<br />

of renal afferent and efferent nerves with resultant decrease<br />

in sympathetic outflow to the kidneys should reduce renin<br />

release and sodium retention, increase renal blood flow, and<br />

lower blood pressure. One-year follow-up data of The Symplicity<br />

HTN-2 trial demonstrates two points: (1) that the<br />

initial significant lowering of blood pressure achieved by the<br />

RDN procedure was maintained at the end of one year and<br />

no procedure-related side effect was revealed by that time;<br />

and (2) the control patients maintained on medical therapy<br />

and whose blood pressure was not adequately controlled<br />

were now permitted to switch over to RDN procedure. These<br />

patients again showed significant drop in blood pressure,<br />

which was maintained at the end of six months. Renal<br />

sympathetic Denervation by radiofrequency ablation thus<br />

may provide a safe and effective adjunctive therapy for<br />

treatment-resistant hypertensive patients.<br />

Contributed by:<br />

Arup Dasbiswas<br />

Professor and HOD, Department of Cardiology,<br />

NRS Medical College, Kolkata, India<br />

E-mail address: arup.dasbiswas@gmail.com


indian heart journal 65 (<strong>2013</strong>) 132e136<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Editorial<br />

Yoga e an ancient solution to a modern epidemic. Ready for<br />

prime time?<br />

Harinder K. Bali<br />

Director, Department of Cardiology, Fortis Hospital, Mohali 160062, Punjab, India<br />

Cardiovascular disease (CVD), the leading cause of morbidity<br />

and mortality worldwide, is clearly of pressing clinical and<br />

economic significance, underscoring the need for effective<br />

primary prevention. 1 Although genetic predisposition plays<br />

an important role, the main culprit responsible for the burgeoning<br />

epidemic of CVD are the modifiable risk factors<br />

related to lifestyle. They not only aid in the initiation but also<br />

in the progression and complications of atherosclerosisrelated<br />

diseases. Hypertension, dyslipidemia and diabetes<br />

are all lifestyle-related conditions and are, therefore, modifiable.<br />

Metabolic syndrome has recently been recognized as an<br />

important risk factor particularly in the <strong>Indian</strong> subcontinent.<br />

Increased sympathetic activity, enhanced cardiovascular<br />

reactivity and reduced parasympathetic tone have been<br />

strongly implicated in the pathogenesis of metabolic syndrome<br />

and in the development and progression of atherosclerosis<br />

and cardiovascular disease. Recent research offers<br />

compelling evidence that chronic psychological stress and<br />

negative affective states contribute significantly to the pathogenesis<br />

and progression of insulin resistance, glucose<br />

intolerance, hypertension, dyslipidemia and other metabolic<br />

syndrome-related conditions and ultimately increase the risk<br />

for CVD morbidity and mortality.<br />

In the light of evidence of strong influence of psychosocial<br />

factors on the development of CVD, mind-body therapies<br />

may have considerable potential in its prevention and<br />

treatment. Of particular interest in this regard is Yoga, an<br />

ancient mind-body discipline which originated in our country<br />

almost 4000 years ago and has been widely used in the<br />

management of hypertension, diabetes and related chronic<br />

insulin resistance conditions. Of the seven major branches of<br />

yoga, Hatha (or forceful), Raja (or classical) and Mantra yoga<br />

are the best known and most widely practiced forms. Both<br />

Hatha and Raja yoga emphasize specific postures (asanas),<br />

including both active and relaxation poses as well as breath<br />

control (pranayama), concentration (dharanas) and meditation<br />

(dhyana).<br />

Yoga is a safe, simple-to-learn, noninvasive and inexpensive<br />

practice requiring little in way of equipment or professional<br />

training. However, as with most practices originating<br />

from the east, it was initially met with a lot of reluctance and<br />

resistance in Western countries. Moreover, lack of systemic<br />

studies did not help its cause. This has changed steadily in the<br />

last few decades with a growing body of evidence 2e6 suggesting<br />

that practice of yoga may reduce risk factors for CVD and<br />

may attenuate signs, reduce complications and improve the<br />

prognosis of those with frank or underlying disease.<br />

Since 1970, more than 50 studies from different countries<br />

have been published in peer reviewed journals investigating<br />

the potential influence of yoga and yoga-based programs on<br />

one or more core indices of CVD including measures of insulin<br />

resistance, lipid profiles, body weight and composition and on<br />

blood pressure. Interventions ranged in length from 40 days to<br />

12 months in various studies and they were carried out on<br />

healthy young adults as well as in patients with type II diabetes,<br />

hypertension and coronary artery disease.<br />

1. Glucose intolerance and diabetes<br />

Most of the studies in patients with glucose intolerance and<br />

frank diabetes have shown significant improvement postintervention<br />

in indices of insulin resistance relative to baseline<br />

values. Overall yoga practice was associated with 5.4%e<br />

33.4% reduction in fasting glucose, 24.5%e27% reduction in<br />

post-prandial glucose and 13.3%e27.3% reduction in glycohemoglobin<br />

with the percentage varying by study population<br />

and design. 7 In the present issue of the journal, Shantakumari<br />

et al 8 have reported the results of their study of effect of yoga<br />

on lipids in diabetic patients. Although it is a well-conducted<br />

randomized study, it is limited by the fact that during three<br />

months of yogic intervention, subjects were under supervision<br />

for only the initial 2 weeks raising concerns about<br />

compliance to the yogic protocol during the unsupervised<br />

E-mail address: hkbalipgi@gmail.com.<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.002


indian heart journal 65 (<strong>2013</strong>) 132e136 133<br />

period. Despite this limitation, the results are encouraging<br />

and consistent with other studies 7 on similar subject. Yogic<br />

intervention showed significant improvement in body weight<br />

and also in total cholesterol, LDL, triglycerides and nonsignificant<br />

elevation in HDL level in the intervention group as<br />

compared to controls.<br />

Although the evidence for beneficial effect has been<br />

observed consistently across studies, these results are<br />

tempered by the fact that most of the studies did not include<br />

control subjects.<br />

2. Lipid profile<br />

Practice of yoga and yoga-based programs may improve lipid<br />

profile in healthy adults and in patients with hypertension,<br />

diabetes and coronary artery disease. Four uncontrolled<br />

studies 3,9e11 and three controlled non-randomized<br />

studies 6,12,13 that ranged from six weeks to 12 months in<br />

duration showed significant improvement in lipid profile<br />

(reduction in total cholesterol, low density lipoproteins and<br />

triglyceride values and increase in HDL cholesterol) over control<br />

values and in comparison to control group as a result of<br />

yoga-based interventions. Several RCTs investigating the effect<br />

of yoga in combination with diet, 6,14,15 education, 16 stress<br />

management 6,16,17 and other therapies 6,14,17 have likewise<br />

demonstrated significant improvement in lipid profiles relative<br />

to controls receiving enhanced usual care, exercise and/or<br />

dietary interventions. Of the 5 RCTs identified in adults with<br />

hypertension, CVD or risk factors for CVD, three 14e16 documented<br />

improvement in all indices of dyslipidemia examined<br />

and one 17 reported significantly greater reduction in triglycerides<br />

but not in cholesterol or LDL. Patel et al 16 studied the<br />

long-term effect of yogic intervention on lipids and concluded<br />

that improvement in lipid profile persisted at 8 months but not<br />

at 4 years. Of those studies demonstrating positive effect, yoga<br />

practice was associated with 5.8%e25.2% decrease in total<br />

cholesterol, 22.0%e28.5% decrease in triglycerides and a<br />

12.8%e26.0% in LDL reduction. 7 The magnitude of the difference<br />

varies from study population and design.<br />

3. Body weight and composition<br />

Body weight and composition is uniformly improved by yogic<br />

interventions. Five RCTs 14,15,17e19 demonstrated improvement<br />

in body weight and/or composition relative to usual care, 14,17,19<br />

diet and exercise 15 and no intervention 18 controls. Studies<br />

reporting improvement in anthropometric indices included<br />

investigations of healthy individuals, 9,13,18,20e23 as well as those<br />

with hypertension, 19 and/or other CVD risk factors, 15 coronary<br />

artery disease 6,14,17 and diabetes. 24,25 In these studies yogic<br />

intervention was associated with 1.5%e13.6% improvement in<br />

body weight. 7<br />

4. Blood pressure<br />

A number of studies including seven uncontrolled studies of<br />

healthy 10,22,26 individuals and hypertensive 11,27e29 adults and<br />

eleven RCTs of healthy 30e33 adults and patients with hypertension<br />

or other CVD risk factors 16,19,34e38 have reported<br />

consistent beneficial effect of even relatively short-term<br />

practice of yoga and yoga-based programs on blood pressure.<br />

The duration of yogic intervention studied in these studies has<br />

ranged from 3 weeks to 6 months. Overall, these studies<br />

demonstrated a 4.6%e24.3% decline in diastolic blood pressure<br />

and 2.6%e21.3% decline in systolic blood pressure with<br />

yoga. The magnitude of decline in blood pressure varied with<br />

the study design and sample population. In a cross-sectional<br />

study of healthy mid-life men with similar lifestyle characteristics,<br />

Vyas et al found that those with both short term and<br />

long term experience in Raja yoga meditation had reduced<br />

diastolic blood pressure as compared to those naïve to<br />

meditation. 39<br />

5. Procoagulant changes and oxidative<br />

stress<br />

Procoagulant changes and damage caused by oxidative stress<br />

have a pivotal role to play in the causation of metabolic syndrome<br />

and in the development and progression of diabetes and<br />

CVD. 40 In an uncontrolled study, it was observed that four<br />

months yogic intervention caused a significant decline in<br />

fibrinogen and an increase in fibrinolytic activity. 41 In a nonrandomized<br />

controlled study of healthy German adults,<br />

Schmidt et al 13 documented a significant fall in fibrinogen<br />

among participants completing a 3-month residential Kriya<br />

yoga program relative to community controls matched on age,<br />

gender, and baseline fibrinogen levels. These trials suggest that<br />

yoga may foster beneficial changes in the coagulation and<br />

fibrinolytic systems in healthy adults. Results of some small<br />

studies 3,13,17,42e44 provide evidence that it may reduce oxidative<br />

stress in both healthy populations and those with chronic<br />

insulin resistance related disorders. Observed changes in other<br />

oxidative stress indices include increase in antioxidants 44 and<br />

antioxidative enzymes, 44 and reductions in free radicals. 43<br />

6. Stress<br />

There is a strong experimental evidence to suggest that yoga<br />

can lead to improvement in both cardiovascular response to<br />

stress and cardiovascular recovery from stress. Cardiovascular<br />

reactivity to stress is strongly associated with insulin<br />

resistance. 45 In addition, it is a major independent predictor of<br />

hypertension, stroke, myocardial infarction and cardiovascular<br />

mortality. 46 Cardiovascular recovery from stress similarly<br />

has been strongly associated with CVD risk. 47<br />

7. Other effects<br />

Population based studies 48e54 have shown that participation<br />

in yoga and yoga-based programs promotes significant<br />

reduction in respiratory rate, heart rate, cortisol concentration,<br />

catecholamine levels, renin activity, and accelerated recovery<br />

time from stress as well as significant improvement in<br />

heart rate variability and baroreceptor sensitivity in both


134<br />

indian heart journal 65 (<strong>2013</strong>) 132e136<br />

healthy 48e53 and hypertensive 54 populations. Collectively,<br />

these studies prove that even short-term practice of yoga may<br />

produce marked reduction in sympathoadrenal activation,<br />

enhance cardio-vagal tone and promote sympatho-vagal<br />

balance.<br />

8. CVD clinical end-points<br />

Large studies that have directly studied the effect of yogic<br />

interventions on clinical end-points of cardiovascular disease<br />

in populations suggest that yoga and yoga-based programs<br />

may attenuate signs, reduce complications and improve the<br />

prognosis of those with frank or underlying disease. In an RCT<br />

of <strong>Indian</strong> men with coronary artery disease, 14 those enrolled<br />

in a 12-month comprehensive yoga program showed retardation<br />

of coronary atherosclerosis, increased regression<br />

and reduced progression of vascular lesions, and reduced<br />

anginal episodes relative to usual care controls. In a study<br />

amongst American seniors, 34 those completing a 12-month<br />

comprehensive yoga-based program demonstrated a decline<br />

in carotid intimal media thickness, an indicator of carotid<br />

atherosclerosis, relative to those receiving usual care or a<br />

comprehensive medical, diet, and exercise intervention. The<br />

decline was correlated inversely with adherence, suggesting a<br />

direct relation between the practice of this program and<br />

atherosclerotic change. There is also some evidence that the<br />

clinical benefits observed following yoga-based programs<br />

might persist long term. In a 4-year follow-up of an earlier RCT<br />

in hypertensive adults, Patel et al 16 found that those who had<br />

participated in an 8-week comprehensive yoga relaxation<br />

program were less likely than usual care controls to be<br />

receiving treatment for CVD related complications, to have<br />

experienced a serious coronary event, or to have electrocardiographic<br />

evidence of ischemia.<br />

significant issues which need to be addressed particularly<br />

about the methodology of many of these trials.<br />

Interpretation of many existing studies is limited by small<br />

sample sizes, lack of appropriate control groups, inadequate<br />

description of methods, selection bias, failure to adjust for<br />

lifestyle characteristics and other potential confounders,<br />

exposure to multiple interventions, inadequacies in statistical<br />

analysis and presentation, or other methodological problems.<br />

In many of the controlled trials, treatment allocation was not<br />

randomized, and direct statistical comparisons were not<br />

made to control groups, potentially biasing the findings. In<br />

addition, the large variation in the nature, duration, intensity,<br />

and delivery methods of the yoga-based interventions used,<br />

even among studies using yoga practice alone, renders comparison<br />

across studies difficult. Therefore, although existing<br />

RCTs have yielded results consistent overall with those of<br />

non-randomized and uncontrolled studies, additional high<br />

quality RCTs are warranted.<br />

11. Present state and future direction<br />

Yoga is an inexpensive method of addressing the major<br />

epidemic of CVD sweeping our country. It has no known side<br />

effects, requires no infrastructure or maintenance charges<br />

and can easily be practiced even by the chronically ill and the<br />

elderly. Time has come for it to be included regularly in our<br />

prescription for primary and secondary prevention of CVD. It<br />

is likely to be even more effective when combined with other<br />

lifestyle measures like exercise and dietary modification. It<br />

should be inculcated in the school curriculum for long-term<br />

benefit and various community centers should offer free yoga<br />

classes to the community. Large multicenter multinational<br />

scientifically conducted studies will go a long way in establishing<br />

its definitive role in CVD prevention and treatment.<br />

9. Possible mechanisms of action of yoga<br />

Although the mechanisms underlying the putative beneficial<br />

effects of yoga therapy on cardiovascular risk profiles are not<br />

yet well understood, the observed changes probably occur primarily<br />

through two pathways. First, by reducing the activation<br />

and reactivity of the sympatho-adrenal system and the hypothalamic<br />

pituitary adrenal (HPA) axis and promoting feelings of<br />

well-being, yoga may alleviate the effects of stress and foster<br />

multiple positive downstream effects on neuroendocrine status,<br />

metabolic function and related inflammatory responses.<br />

Second, by directly stimulating the vagus nerve, yoga may<br />

enhance parasympathetic output and thereby shift the autonomic<br />

nervous system balance from primarily sympathetic to<br />

parasympathetic leading to positive changes in cardiac-vagal<br />

function, in mood and energy state, and in related neuroendocrine,<br />

metabolic, and inflammatory responses.<br />

10. Limitations of studies<br />

Although there is a wealth of data available to support the<br />

efficacy of yoga and yoga related interventions, there are<br />

references<br />

1. American <strong>Heart</strong> Association. 2012 <strong>Heart</strong> and Stroke Statistical<br />

Update. American <strong>Heart</strong> Association; 2012.<br />

2. Pandya D, Vyas V, Vyas S. Mind-body therapy in the<br />

management and prevention of coronary disease. Compr Ther.<br />

1999;25:283e293.<br />

3. Damodaran A, Malathi A, Patil N, Shah N, Suryavansihi<br />

<strong>Mar</strong>athe S. Therapeutic potential of yoga practices in<br />

modifying cardiovascular risk profile in middle aged men<br />

and women. J Assoc Physicians India. 2002;50:633e640.<br />

4. Sahay B, Sahay R. Lifestyle modification in man e agement of<br />

diabetes mellitus. J <strong>Indian</strong> Med Assoc. 2002;100:178e180.<br />

5. Raub J. Psychophysiologic effects of hatha yoga on<br />

musculoskeletal and cardiopulmonary function: a literature<br />

review. J Altern Complement Med. 2002;8:797e812.<br />

6. Yogendra J, Yogendra H, Ambardekar S, et al. Beneficial<br />

effects of yoga lifestyle on reversibility of ischaemic heart<br />

disease: Caring <strong>Heart</strong> Project of International Board of Yoga.<br />

J Assoc Physicians India. 2004;52:283e289.<br />

7. Innes KE, Bourguignon C, Taylor AG. Risk indices associated<br />

with the insulin resistance syndrome, cardiovascular disease,<br />

and possible protection with yoga: a systematic review. JAm<br />

Board Fam Pract. 2005;18:491e519.


indian heart journal 65 (<strong>2013</strong>) 132e136 135<br />

8. Shantakumari N, Sequeria S, Eldeep Rasha. Effects of a yoga<br />

intervention on lipid profiles of diabetes patients with<br />

dyslipidemia. <strong>Indian</strong> <strong>Heart</strong> J. <strong>2013</strong>;65:127e131.<br />

9. Udupa KN, Singh RH. The scientific basis of yoga. JAMA.<br />

1972;220:1365.<br />

10. Joseph S, Sridharan K, Patil S, et al. Study of some<br />

physiological and biochemical parameters in subjects<br />

undergoing yogic training. <strong>Indian</strong> J Med Res.<br />

1981;74:120e124.<br />

11. Patel C. Reduction of serum cholesterol and blood pressure in<br />

hypertensive patients by behaviour modification. J Roy Coll<br />

Gen Pract. 1976;26:211e215.<br />

12. Naruka J, Mathur R, Mathur A. Effect of pranayama practices<br />

on fasting blood glucose and serum cholesterol. <strong>Indian</strong> J Med<br />

Sci. 1986;40:149e152.<br />

13. Schmidt T, Wijga A, Muhlen Von Zur, et al. Changes in<br />

cardiovascular risk factors and hormones during a<br />

comprehensive residential three month kriya yoga training<br />

and vegetarian nutrition. Acta Physiol Scand Suppl.<br />

1997;640:158e162.<br />

14. Manchanda S, Narang R, Reddy K, et al. Retardation of<br />

coronary atherosclerosis with yoga lifestyle intervention.<br />

J Assoc Physicians India. 2000;48:687e694.<br />

15. Mahajan A, Reddy K, Sachdeva U. Lipid profile of coronary<br />

risk subjects following yogic lifestyle intervention. <strong>Indian</strong><br />

<strong>Heart</strong> J. 1999;51:37e40. 100.<br />

16. Patel C, <strong>Mar</strong>mot M, Terry D, et al. Trial of relaxation in<br />

reducing coronary risk: four year follow-up. Br Med J.<br />

1985;290:1103e1106.<br />

17. Jatuporn S, Sangwatanaroj S, Saengsiri A, et al. Short-term<br />

effects of an intensive lifestyle modification program on lipid<br />

peroxidation and antioxidant systems in patients with<br />

coronary artery disease. Clin Hemorheol Microcirc.<br />

2003;29:429e436.<br />

18. Bera TK, Rajapurkar MV. Body composition, cardiovascular<br />

endurance and anaerobic power of yogic practitioner. <strong>Indian</strong> J<br />

Physiol Pharmacol. 1993;37:225e228.<br />

19. Murugesan R, Govindarajulu N, Bera T. Effect of selected yogic<br />

practices on the management of hypertension. <strong>Indian</strong> J Physiol<br />

Pharmacol. 2000;44:207e210.<br />

20. Udupa KN, Singh RH, Settiwar RM. Physiological and<br />

biochemical studies on the effect of yogic and certain other<br />

exercises. <strong>Indian</strong> J Med Res. 1975;63:620e624.<br />

21. Satyanarayana M, Rajeswari KR, Rani NJ, Krishna CS, Rao PV.<br />

Effect of santhi kriya on certain psychophysiological<br />

parameters: a preliminary study. <strong>Indian</strong> J Physiol Pharmacol.<br />

1992;36:88e92.<br />

22. Telles S, Nagarathna R, Nagendra HR, Desiraju T.<br />

Physiological changes in sports teachers following 3 months<br />

of training in yoga. <strong>Indian</strong> J Med Sci. 1993;47:235e238.<br />

23. Raju PS, Prasad KV, Venkata RY, Murthy KJ, Reddy MV.<br />

Influence of intensive yoga training on physiological changes<br />

in 6 adult women: a case report. J Altern Complement Med.<br />

1997;3:291e295.<br />

24. Divekar M, Bhat M, Mulla A. Effect of yoga therapy in diabetes<br />

and obesity. J Diab Assoc Ind. 1978;17:75e78.<br />

25. Jain S, Uppal A, Bhatnagar S, Talukdar B. A study of response<br />

pattern of non-insulin dependent diabetics to yoga therapy.<br />

Diabetes Res Clin Pract. 1993;19:69e74.<br />

26. Anantharaman R, Kabir R. A study of yoga. J Psychol Res.<br />

1984;28:97e101.<br />

27. Mogra A, Singh G. Effect of biofeedback and yogic<br />

relaxation exercise on the blood pressure levels of<br />

hypertensives: a preliminary study. Aviation Med.<br />

1986;30:68e75.<br />

28. Lakshmikanthan C, Alagesan R, Thanikachalam S, et al.<br />

Long term effects of yoga on hypertension and/or coronary<br />

artery disease. J Assoc Physicians India. 1979;27:1055e1058.<br />

29. Sundar S, Agrawal S, Singh V, Bhattacharya S, Udupa K,<br />

Vaish S. Role of yoga in management of essential<br />

hypertension. Acta Cardiol. 1984;39:203e208.<br />

30. Bagga OP, Gandhi A. A comparative study of the effect<br />

of Transcendental Meditation (T.M.) and shavasana<br />

practice on cardiovascular system. <strong>Indian</strong> <strong>Heart</strong> J.<br />

1983;35:39e45.<br />

31. Cusumano JA, Robinson SE. The short-term<br />

psychophysiological effects of hatha yoga and progressive<br />

relaxation on female Japanese students. Appl Psychol.<br />

1992;42:77e90.<br />

32. Ray U, Mukhopadhyaya S, Purkayastha S, et al. Effect of<br />

yogic exercises on physical and mental health of young<br />

fellowship course trainees. <strong>Indian</strong> J Physiol Pharmacol.<br />

2001;45:37e53.<br />

33. Harinath K, Malhotra AS, Pal K, et al. Effects of hatha yoga<br />

and omkar meditation on cardiorespiratory performance,<br />

psychologic profile, and melatonin secretion. J Altern<br />

Complement Med. 2004;10:261e268.<br />

34. Fields JZ, Walton KG, Schneider RH, et al. Effect of a<br />

multimodality natural medicine program on carotid<br />

atherosclerosis in older subjects: a pilot trial of Maharishi<br />

Vedic Medicine. Am J Cardiol. 2002;89:952e958.<br />

35. Patel C, <strong>Mar</strong>mot MG, Terry DJ. Controlled trial of biofeedbackaided<br />

behavioural methods in reducing mild hypertension. Br<br />

Med J (Clin Res Ed). 1981;282:2005e2008.<br />

36. Patel C, <strong>Mar</strong>mot M. Can general practitioners use training in<br />

relaxation and management of stress to reduce mild<br />

hypertension? Br Med J (Clin Res Ed). 1988;296:21e24.<br />

37. Patel C, North WR. Randomised controlled trial of yoga and<br />

bio-feedback in management of hypertension. Lancet.<br />

1975;2:93e95.<br />

38. Broota A, Varma R, Singh A. Role of relaxation in<br />

hypertension. J <strong>Indian</strong> Acad Appl Psychol. 1995;21:29e36.<br />

39. Vyas R, Dikshit N. Effect of meditation on respiratory system,<br />

cardiovascular system and lipid profile. <strong>Indian</strong> J Physiol<br />

Pharmacol. 2002;46:487e491.<br />

40. Ceriello A, Motz E. Is oxidative stress the pathogenic<br />

mechanism underlying insulin resistance, diabetes and<br />

cardiovascular disease? the common soil hypothesis<br />

revisited. Arterioscler Thromb Vasc Biol. 2004;24:816e823.<br />

41. Chohan IS, Nayar HS, Thomas P, Geetha NS. Influence of<br />

yoga on blood coagulation. Thromb Hemost. 1984;51:196e197.<br />

42. Singh S, Malhotra V, Singh K, Sharma S. A pre-liminary<br />

report on the role of yoga asanas on oxi-dative stress in noninsulin<br />

dependent diabetes. <strong>Indian</strong> J Clin Biochem.<br />

2001;16:216e220.<br />

43. Bhattacharya S, Pandey U, Verma N. Improvement in<br />

oxidative status with yogic breathing in young healthy males.<br />

<strong>Indian</strong> J Physiol Pharmacol. 2002;46:349e354.<br />

44. Sharma H, Sen S, Singh A, Bhardwaj NK, Kochupillai V,<br />

Singh N. Sudarshan kriya practitioners exhibit better<br />

antioxidant status and lower blood lactate levels. Biol Psychol.<br />

2003;63:281e291.<br />

45. Moan A, Nordby G, Rostrup M, Eide I, Kjeldsen SE.<br />

Insulin sensitivity, sympathetic activity, and<br />

cardiovascular reactivity in young men. Am J Hy-pertens.<br />

1995;8:268e275.<br />

46. Jennings JR, van der Molen MW, Somsen RJ, Graham R,<br />

Gianaros PJ. Vagal function in health and disease: studies in<br />

Pittsburgh. Physiol Behav. 2002;77:693e698.<br />

47. Mezzacappa ES, Kelsey RM, Katkin ES, Sloan RP. Vagal<br />

rebound and recovery from psychological stress. Psychosom<br />

Med. 2001;63:650e657.<br />

48. Bernardi L, Sleight P, Bandinelli G, et al. Effect of<br />

rosary prayer and yoga mantras on autonomic<br />

cardiovascular rhythms: comparative study. BMJ.<br />

2001;323:1446e1449.


136<br />

indian heart journal 65 (<strong>2013</strong>) 132e136<br />

49. Konar D, Latha R, Bhuvaneswaran JS. Cardiovascular<br />

responses to head down-body-up postural ex-ercise<br />

(sarvangasana). <strong>Indian</strong> J Physiol Pharmacol. 2000;44:392e400.<br />

50. Bowman A, Clayton R, Murray A, Reed J, Subhan M, Ford G.<br />

Effects of aerobic exercise training and yoga on the baroreflex<br />

in healthy elderly persons. Eur J Clin Invest. 1997;27:443e449.<br />

51. Udupa K, Madanmohan, Bhavanani AB, Vijayalakshmi P,<br />

Krishnamurthy N. Effect of pranayam training on cardiac<br />

function in normal young volunteers. <strong>Indian</strong> J Physiol<br />

Pharmacol. 2003;47:27e33.<br />

52. Vempati R, Telles S. Baseline occupational stress<br />

levels and physiological responses to a two day<br />

stress management program. J<strong>Indian</strong>Psychol. 2000;<br />

18:33e37.<br />

53. Vempati R, Telles S. Yoga-based guided relaxation reduces<br />

sympathetic activity judged from baseline levels. Psychol Rep.<br />

2002;90:487e494.<br />

54. Selvamurthy W, Sridharan K, Ray U, et al. A new<br />

physiological approach to control essential hypertension.<br />

<strong>Indian</strong> J Physiol Pharmacol. 1998;42:205e213.


indian heart journal 65 (<strong>2013</strong>) 201e218<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

How Do I Do It<br />

Stepwise evaluation of left to right shunts by<br />

echocardiography<br />

Neeraj Awasthy a , S. Radhakrishnan b, *<br />

a Associate Consultant, Department of Pediatric and Congenital <strong>Heart</strong> Diseases, Fortis Escorts <strong>Heart</strong> Institute, Delhi, India<br />

b Director, Department of Pediatric and Congenital <strong>Heart</strong> Diseases, Fortis Escorts <strong>Heart</strong> Institute, Delhi, India<br />

Echocardiography has revolutionized the practice of pediatric<br />

cardiology. The addition of Doppler and color flow mapping<br />

also gives physiological information about flow and pressures<br />

and enables the pediatric cardiologist to refer patients for<br />

surgical treatment without cardiac catheterization, especially<br />

in neonate and infants. In this communication echocardiographic<br />

findings of common shunt lesions are discussed.<br />

1. General features: shunt lesions<br />

arteries or obstruction in pulmonary vascular bed (as in<br />

pulmonary arterial hypertension).<br />

- Increase in pressures in RV and beyond may be seen in<br />

large VSD, similarly a large PDA would lead to significant<br />

increase in PA pressures<br />

4) The magnitude of the gradient from a chamber outflow<br />

would be dependent on the magnitude of shunt into the<br />

chamber.<br />

- This may lead to exaggerated gradients even in hemodynamically<br />

scuttle lesions viz. exaggerated pulmonary<br />

There are a few salient features of all the shunt lesions:<br />

1) The shunt would lead to volume overloads of the chambers<br />

it feeds (particularly in relation to the tricuspid<br />

valves) e.g. while a shunt proximal to the tricuspid valve<br />

would lead to volume overloading of the right atrium and<br />

right ventricle and further (Fig. 1). A lesion beyond the<br />

tricuspid valve would lead to the volume overloading of<br />

the left atrium (LA) and left ventricle (LV).<br />

2) The magnitude of the chamber enlargement depends<br />

upon the magnitude of the shunt. Thus significant right<br />

atrium (RA) and right ventricle (RV) enlargement would be<br />

a feature of pretricuspid shunt, while a significant LA and<br />

LV enlargement would be a feature of post-tricuspid<br />

shunt.<br />

3) The pressure of the investigated chamber would rise not<br />

only on account of distal obstruction (obstruction of the<br />

outflow of the chamber) or it would be because of the<br />

transmitted pressures from the adjacent chambers on<br />

account of the shunt.<br />

- Increase in RV pressures may be because of distal pulmonary<br />

stenosis, obstruction in branch pulmonary<br />

Fig. 1 e 4 Chamber view showing prominent right atrium<br />

and right ventricle. The image has been taken from a case<br />

of 15-year-old child with large atrial septal defect.<br />

* Corresponding author. Tel.: þ91 (0) 9811962775.<br />

E-mail addresses: n_awasthy@yahoo.com, n_awasthy@yahoomail.com (N. Awasthy), samurai43@yahoo.com (S. Radhakrishnan).<br />

0019-4832/$ e see front matter Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.003


202<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

Fig. 2 e Perimembranous defect shown with anterior tilt<br />

from 4c view.<br />

stenosis gradients in associated pretricuspid shunts<br />

(like ASD), exaggerated mitral and aortic valve gradients<br />

in associated post-tricuspid shunts like VSD 1 or PDA in<br />

absence of significant stenosis. For example gradients<br />

upto 60 mmHg have been reported across pulmonary<br />

valve in patients of atrial septal defect in absence of<br />

pulmonary stenosis (Fig. 2).<br />

5) The secondary manifestations of the shunt lesions may<br />

themselves lead to exaggerated secondary effects e.g. the<br />

mitral annular dilatation on account of post-tricuspid<br />

shunt may lead to mitral regurgitation and this may<br />

further lead to mitral valve dilatation and LV dilatation.<br />

6) Since a shunt lesion almost always signifies a communication<br />

between 2 chamber the gradient between the 2<br />

chambers can guide as to the magnitude of the shunt<br />

lesion or the size of the defect (Fig. 3A and B).<br />

7) The size of the defect in 2 dimensions may be a useful<br />

guide in deciding the degree of shunt. It also is useful to<br />

help the interventional modality<br />

- The size of the VSD may be compared to the size of the<br />

Aortic root for classifying the size of the VSD.<br />

- The size of defects like ASD, coronary AV fistulae, VSD,<br />

RSOV etc would help in deciding the size of the device<br />

which may be used.<br />

Fig. 3 e Echocardiographic imaging with continuous wave Doppler gradient across the PDA, showing the PDA gradient of<br />

89 mmHg against systemic pressures of 100 mmHg. The PA pressures from the gradient is systemic pressures (100 mmHg)<br />

PDA gradient (89 mmHg) [ 11 mmHg. B shows the gradient across the VSD of 98 mmHg against systemic pressures of<br />

120 mmHg. By the observation the predicted RV pressures are 22 mmHg (100e98).


indian heart journal 65 (<strong>2013</strong>) 201e218 203<br />

Fig. 4 e Transesophageal echocardiography showing the dimensions of ASD in various TEE planes. The view is obtained by<br />

keeping the endoscope in the middle of the esophagus and rotating the icon four chamber view, A: (showing atrial and<br />

atrioventricular valve rims), Basal short axis view, B: shows the atrial and aortic rims, basal long axis view, C: shows the<br />

superior vena cava (SVC) and inferior vena cava (IVC) rims. For optimum device placement the rims should be adequate (at<br />

least 5 mm) and supportive. The size of the ASD has to be seen in all the planes to decide the size of the ASD device. The<br />

same (D) has to be viewed on color Doppler to ascertain the flow and the visualization of additional defect.<br />

8) Echocardiography should focus, not only on the characteristics<br />

of the primary lesion, but also on the adjacent<br />

structures of the defect e.g. distances from the adjoining<br />

valves<br />

- VSD, it is important to note the distances from the aortic<br />

valve when considering for device closure. It is also<br />

important from the surgical point of view.<br />

Fig. 5 e An interesting case of ALCAPA with masked manifestations. In view of associated large VSD leading to pulmonary<br />

artery hypertension the flow in the anomalous coronary artery from the pulmonary artery on color flow was normal (A). Thus<br />

2d echocardiography (B) becomes important in such cases demonstrating the origin of the left main coronary artery (LMCA)<br />

from pulmonary artery (PA).


204<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

Fig. 6 e An interesting case of coronary sinus type of ASD with mitral stenosis and unroofed ASD. The interesting case<br />

demonstrated that because of associated large coronary sinus ASD (A), mitral gradients were underestimated on color<br />

Doppler echocardiography (B) with mean gradient of 3 mmHg inspite of severe mitral stenosis (mitral valve area on


indian heart journal 65 (<strong>2013</strong>) 201e218 205<br />

Table 1 e Stepwise evaluation for ASD.<br />

1) Initial indication is the volume overload of the chambers (RA<br />

and RV) in 4c view<br />

2) Visualize the defect from subcostal view (Sagittal and coronal)<br />

3) Determine the site of defect: fossa ovalis or others (others being<br />

not suitable for device)<br />

4) Determine the direction of shunting of the defect<br />

5) Look for the associated structures particularly pulmonary veins<br />

and AV valves<br />

6) Look for the pulmonary artery pressures: TR and PR gradients<br />

7) Determine suitability for device closure<br />

Table 3 e Stepwise evaluation for a PDA.<br />

1) Visualize the ductus ostium and aortic isthmus from the parasternal<br />

short axis, high parasternal short axis and suprasternal<br />

views<br />

2) Determine the direction of shunt by color flow mapping and<br />

Doppler<br />

3) Take the peak velocity of the PDA signal which will give the<br />

pressure difference between the aorta and pulmonary artery<br />

and obtain the aortic, RVOT and PA velocities<br />

4) Take the measurements of the left ventricle and left atrium as<br />

these will reflect the volume of the left to right shunt<br />

5) Specifically look for associated defects like coarctation of aorta<br />

(suprasternal view), aortic interruption and aortopulmonary<br />

window (communication between the ascending aorta and<br />

pulmonary artery)<br />

Table 2 e Stepwise evaluation for VSD.<br />

1) Initial indication is the volume overload of the chambers (LA<br />

and LV)<br />

2) Visualize the defect from all possible windows to look for the<br />

defect<br />

3) Determine the presence of additional defects (screening the<br />

septum e septal sweep in subcostal view, apical 4c view and<br />

parasternal short and long axis sweep in color and 2d)<br />

4) Determine the direction of shunting of the defect<br />

5) Look for the associated structures particularly outflows<br />

6) Look for the pulmonary artery pressures: VSD gradients, TR and<br />

PR gradients<br />

7) Determine the volume overload of chambers (Z scores of LV,<br />

particularly in M mode)<br />

8) Determine suitability for device closure<br />

- For ASD, the rims are seen not only for their adequacy,<br />

but also the adjoining structures being encroached<br />

upon, whenever contemplating a device closure<br />

(Fig. 4AeD).<br />

- For AP window, the distance from coronaries and valves<br />

becomes important<br />

- The post-tricuspid shunt is known to mask the manifestations<br />

of Anomalous left coronary artery from pulmonary<br />

artery (ALCAPA), and thus one should keenly<br />

look at the 2d anatomy and origin of the coronary<br />

arteries whenever investigating an associated shunt<br />

lesion 2,3 (Fig. 5A and B).<br />

9) Whenever investigating a shunt at multiple sites or an<br />

associated lesion, one must remember that the shunt flow<br />

may be modified by the presence of distal obstruction and<br />

also by the associated shunt.<br />

- The associated post-tricuspid shunt may lead to exaggerated<br />

manifestations of a pretricuspid shunt lesions<br />

(viz ASD). Thus RA and RV may be unduly dilated even in<br />

the presence of small ASD, with the associated presence<br />

of post-tricuspid shunt (VSD or PDA) or associated<br />

presence of mitral stenosis.<br />

- The associated aortic stenosis or coarctation of aorta<br />

may exaggerate the shunt across the ventricular septal<br />

defect.<br />

10) The associated lesions being drained off by the shunt lesions<br />

may become masked and may manifest themselves<br />

only after the shunt lesion is closed.<br />

- Mitral stenosis may not manifest itself in the presence<br />

of ASD (although it may exaggerate the shunt flow<br />

across it) (Fig. 6AeF).<br />

- The manifestations of significant mitral regurgitation<br />

may get unmasked after ASD closure. 4<br />

- High LVedp may not only exaggerate ASD shunt, they<br />

may manifest themselves as pulmonary edema after<br />

ASD closure.<br />

- VSD or PDA may mask the gradients across the aortic<br />

stenosis or coarctaion of aorta and this may manifest<br />

itself after the treatment of the underlying shunt<br />

lesion. 5<br />

11) Systemic disorders and conditions may exaggerate or<br />

confound the features and even echocardiographic features<br />

of a shunt lesion.<br />

- Anemia may exaggerate the gradients across any shunt<br />

lesion or across valves. Anemia may lead to LV dilatation<br />

thus confounding the assessment of associated<br />

post-tricuspid shunt lesion.<br />

- Systemic hypertension may not only lead to exaggerated<br />

shunt gradients, it may also lead to secondary<br />

ventricular hypertrophy thus leading to high LVedps<br />

and exaggerating ASD shunt.<br />

- Hyperdynamic states such as fever, anemia, thyroid<br />

disorders may exaggerate the shunt gradients.<br />

Assessment of shunt lesions must be preceded by a<br />

complete clinical information, chest X-ray and electrocardiogram.<br />

There would be a situation when information<br />

gained by these investigations will lead to clinical decision<br />

when echocardiographic findings are equivocal.<br />

planimetry of 0.7 cm 2 ) (C). The another interesting hemodynamic aspect of the lesion was associated unroofed coronary<br />

sinus ASD leading to right to left shunting and hence systemic desaturation. This shunting was clearly demonstrated by<br />

contrast injection in left brachial artery (D and E). The injection showed early filling of the left atrium and left ventricle (D)<br />

followed by right atrium and right ventricle (via coronary sinus ASD): (F). The draining of the LSVC to left atrium can be<br />

confirmed by computed tomography scan.


206<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

Fig. 7 e 2d echocardiography with subcostal view showing<br />

bicaval view with SVC and IVC rims and left to right shunt.<br />

1.1. Stepwise approach (on echocardiography)<br />

Stepwise approach (on echocardiography) for evaluation of<br />

any shunt lesion involves:<br />

1) Determine the presence of shunt lesion<br />

2) Determine the volume overload (and complication) on account<br />

of the shunt lesion<br />

3) Defining the size of the lesion and its location<br />

4) Define the lesion on echocardiography for operability<br />

5) If suitable to decide the relevant modality of treatment<br />

The essential approach to any lesion should not be directed<br />

at the shunt lesion, rather it should be a standardized<br />

sequential analysis, as it is not the shunt lesion in isolation<br />

that exist and one needs to evaluate all the structural heart<br />

Fig. 9 e Apical 4 chamber view showing large ostium<br />

primum ASD defect in lower part of the interatrial septum<br />

marked by the arrow.<br />

defects. A few salient features of the important shunt<br />

lesionseASD, VSD, PDA and AP window are illustrated below<br />

Table 1.<br />

1.1.1. Step 1: suspect the shunt lesion<br />

Visualization of an accessory flow into the chamber<br />

Any shunt lesion will lead to the chamber enlargement into<br />

which it drains<br />

Tricuspid valve an important landmark<br />

A pretricuspid shunt would lead to right atrium and right<br />

ventricular chamber enlargement<br />

Fig. 8 e 2d echocardiography with color comparison showing the SVC type of sinus venosus ASD. There is overlapping of<br />

the SVC over the defect with partial anomalous pulmonary venous drainage of right upper pulmonary vein to SVC (PAPVC).


indian heart journal 65 (<strong>2013</strong>) 201e218 207<br />

fistulae and RSOV to any structure beyond tricuspid valve,<br />

aorta e LV tunnel, aortopulmonary collaterals<br />

1.1.3. Step 3: critically see the chamber enlargement<br />

The enlargement of the innominate vein and superior vena<br />

cava (SVC) will point toward a flow into the SVC<br />

Enlargement of the isolated SVC in the absence of innominate<br />

vein dilatation will point toward a shunt in SVC or an AV<br />

malformation draining to SVC<br />

2. Atrial septal defects (Table 1)<br />

2.1. Objectives on echocardiography<br />

Fig. 10 e Transesophageal echocardiography at the level of<br />

basal long axis view showing the characteristics of SVC<br />

type of sinus venosus ASD. There is clearly a defect with<br />

overlying of the SVC over the defect. At times slight<br />

retroflexion of the probe may profile the defect well.<br />

1. To diagnose atrial septal defect, asses its anatomical site<br />

and size. 6e8<br />

2. To assess the direction and quantum of flow.<br />

3. To assess the degree of pulmonary arterial hypertension.<br />

4. To assess atrioventricular valve anomalies, pulmonary<br />

veins and pulmonary valve stenosis.<br />

2.2. ASD classification<br />

A post-tricuspid shunt would lead to left atrial and left<br />

ventricular enlargement<br />

1.1.2. Step 2: a recall of the shunt lesions<br />

Pretricuspid shunts: atrial septal defect (ASD), interatrial<br />

communication, anomalous pulmonary venous drainage,<br />

systemic AV fistulae, Ruptured sinus of Valsalva to right<br />

atrium, Coronary AV fistulae to right atrium, Gerbode defect<br />

(left ventricle to right atrial shunt)<br />

Post-tricuspid shunt: Ventricular septal defect (VSD)<br />

Table 2, aortopulmonary window (APW), Patent ductus arteriosus<br />

(PDA) Table 3, pulmonary AV fistulae, coronary AV<br />

Defects of atrial septum are classified into: a. patent foramen<br />

ovale (PFO), b. fossa ovalis atrial septal defect (Fig. 7), c. sinus<br />

venosus defect (Fig. 8), d. Coronary sinus atrial septa defect<br />

(Fig. 6A, DeF), e. ostium primum atrial septal defect (Fig. 9).<br />

2.3. Evaluation on echocardiography<br />

The abnormal interventricular septal motion and enlarged<br />

right atrium and ventricle are indirect evidence of left to right<br />

shunt at atrial level.<br />

The best views to directly visualize the atrial septal defect<br />

are subcostal coronal and sagittal views.<br />

Fig. 11 e 2d echocardiography with subcostal sagittal view showing the case of total anomalous pulmonary venous<br />

drainage with right to left shunt across atrial septal defect.


208<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

most part of the interatrial septum with atrioventricular<br />

valves attached at the same level are designated as ostium<br />

primum defects.<br />

It should also be viewed in apical four chamber and short<br />

axis views.<br />

The four chamber view will also show the attachments of<br />

the atrioventricular valves. These will be at the same level in<br />

ostium primum defect.<br />

The color flow mapping across the defect will show the<br />

direction of flow and also presence or absence of any<br />

regurgitation.<br />

Doppler velocities across all valves should be taken, in<br />

particular the pulmonary valve to look for any pulmonary<br />

stenosis.<br />

False positive and false negatives<br />

Fig. 12 e Schematic diagram of the interventricular septum<br />

with removed RV cavity from the RV side. Various part of<br />

the septum are profiled: blue (muscular septum) this forms<br />

the region of the muscular VSD. The muscular septum can<br />

further be classified with respect to the muscular<br />

moderator and septal band. Yellow region signifies the<br />

perimembranous region, purple color signifies the inlet<br />

septum and green color the outlet septum.<br />

Atrial septal defect will be diagnosed by a dropout in the<br />

interatrial septum with flow across the defect on Doppler<br />

interrogation.<br />

When the defect is visualized its relationship to the SVC and<br />

inferior vena Cava (IVC) should be evaluated. If the SVC<br />

forms the roof of the defect, the defect is SVC sinus venous<br />

type. If the IVC straddles the defect, it is IVC type. The defects<br />

in the center of the atrial septum involving the fossa<br />

ovalis area are fossa ovalis defects. The defect in the lower<br />

Apical four chamber views are notorious in giving false echo<br />

dropouts and false positive impression of a defect.<br />

Apical four chamber view can completely miss an SVC type<br />

defect because of its very superior location. Transesophageal<br />

echo will resolve this issue (Fig. 10).<br />

Hugely dilated coronary sinus (as in coronary sinus TAPVC)<br />

has been mistaken for ostium primum ASD in inexperienced<br />

hands because of its very posterior location.<br />

The inflow velocities of the AV valves should be seen to rule<br />

out any mitral valve obstruction. The associated mitral<br />

obstruction may get missed unless specifically seen on 2<br />

dimensional echocardiography, as there may not be significant<br />

gradient across the mitral valve even with significant<br />

obstruction because of associated ASD (true for all Lutembacher<br />

cases) (Fig. 6 AeF).<br />

All pulmonary veins should be specifically imaged to see if<br />

they are abnormally connected or not and to look for any<br />

pulmonary vein stenosis. The pulmonary veins are best<br />

seen in subcostal coronal and sagittal, apical four chamber<br />

and short axis and suprasternal short axis views.<br />

Fig. 13 e 2d echocardiography with four chamber view with anterior tilt showing the perimembranous VSD defect getting<br />

restricted by septal leaflet of the tricuspid valve in 2d (A) and color Doppler (B).


indian heart journal 65 (<strong>2013</strong>) 201e218 209<br />

Fig. 14 e 2d echocardiography with subcostal coronal view with anterior tilt with opening of the aortic outflow showing the<br />

outlet muscular VSD with left to right shunt in 2d (14A) and color mapping (B).<br />

3. Direction of shunt<br />

Dominant shunt occurs from left to right. Left to right shunt<br />

occurs mainly during mid to late systole as ‘v’ wave of left atria<br />

is larger than right atria (Fig. 7). 8e11 The second wave of left to<br />

right shunt occur with atrial contraction. The following<br />

pattern of shunting can be appreciated in patient of ASD.<br />

1) Left to right shunt: In associated ASD without pulmonary<br />

artery hypertension (PAH) this is a commonest pattern of<br />

shunting. Onset of atrial fibrillation even in absence of PAH<br />

can cause bidirectional shunt.<br />

2) Right to left shunt: In ASD right to left shunting will occur in<br />

following situation (Fig. 11):<br />

a) Severe PAH.<br />

b) Hypoplasia of right sided chambers which may rarely<br />

be isolated but more often as a part of more complex<br />

anomalies.<br />

c) Severe pulmonary stenosis with right ventricular hypertrophy<br />

or hypertension.<br />

d) Obligatory right to left shunt even in absence of significant<br />

PAH is seen in tricuspid atresia, TAPVC.<br />

3) Bidirectional shunting across atrial septal defect is seen<br />

most commonly during the stage of progression of PAH<br />

For the estimation of pulmonary arterial pressure the peak<br />

gradient of tricuspid regurgitation should be taken. If pulmonary<br />

regurgitation is present the pressure derived from the<br />

peak diastolic velocity will reflect the pulmonary arterial<br />

mean pressure.<br />

3.1. Calculation of shunt in ASD<br />

Because of its fallacy calculation of shunts by echocardiography<br />

is rarely practiced. More often quantitative assessment of<br />

significant shunt is assessed by its impact on right atrial or<br />

right ventricular chamber size. Thus a “significant” shunt is<br />

Fig. 15 e 2d echocardiography with parasternal long axis view showing the apical muscular type of trabecular muscular<br />

VSD with left to right shunt in 2d (A) and color mapping (B).


210<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

Fig. 16 e 2d echocardiography with parasternal long axis view showing the doubly committed VSD with left to right shunt<br />

in 2d (A) and color flow mapping (B).<br />

will be associated with dilated right atrium and ventricle. The<br />

disadvantage of this visual assessment is that progressive<br />

enlargement of RA and RV can occur with increasing pulmonary<br />

artery pressures. Thus hugely dilated right atrium and<br />

ventricle will persist even with Eisenmenger state where there<br />

is only right to left shunt. Diagnostic cardiac catheterization in<br />

atrial septal defects is indicated when required to measure<br />

pulmonary artery pressures or to evaluate pulmonary<br />

vascular resistance.<br />

3.2. Evaluation of atrial septum by transesophageal<br />

echocardiography (Fig. 4AeD)<br />

Views, which are most useful for evaluation of atrial septal<br />

defect on TEE, 6,7,10 are<br />

a. Basal short axis view, b. bicaval or basal long axis view,<br />

c. Four chamber view.<br />

3.2.1. Basal short axis view<br />

Obtained by keeping the endoscope at the middle part of<br />

esophagus.<br />

The aortic and atrial rims can be best seen in this view.<br />

The fossa ovalis defect is seen in middle part of defect while<br />

sinus venosus defect is seen in the upper part of defect.<br />

3.2.2. Basal long axis or bicaval view<br />

Fig. 17 e 2d echocardiography shows in 4 chamber view<br />

shows the large inlet VSD getting partially restricted by<br />

septal leaflet of tricuspid valve. Such partial closure of VSD<br />

is more commonly a feature of perimembranous VSD and<br />

is very rarely seen in the inlet VSD as in the present case.<br />

Endoscope at the same level and rotating the icon to 80e100 0 .<br />

Fossa ovalis defect is seen in middle part of septum, while<br />

sinus venosus defect is seen in relation to superior vena<br />

cava with superior vena cava type of defect or in relation to<br />

inferior vena cava with inferior vena cava type of defect<br />

along with partial anomalous venous drainage of pulmonary<br />

vein.<br />

SVC and IVC rims can be accurately assessed in this view.<br />

3.2.3. Four-chamber view<br />

Obtained by keeping the endoscope at the lower part of<br />

esophagus.


indian heart journal 65 (<strong>2013</strong>) 201e218 211<br />

Atrial and atrioventricular valve rims of fossa ovalis defect<br />

are visualized and volume overloaded right atrium and right<br />

ventricle.<br />

By rotating the endoscope we can see the attachment of<br />

right and left pulmonary veins to left atrium.<br />

4. Ventricular septal defect (Table 2)<br />

4.1. Objectives of echocardiography<br />

Confirm ventricular septal defect (VSD).<br />

Determine the size and morphological location of VSDs.<br />

Rule out associated lesions.<br />

Assessment of chamber size and wall thickness.<br />

Estimation of shunt size (pulmonary/systemic flow ratio).<br />

Estimate right ventricular and pulmonary arterial pressures.<br />

4.2. Classification of VSD (Fig. 12)<br />

Ventricular septal defects can be classified into following<br />

types 12,13 : a e perimembranous ventricular septal defect<br />

(Figs. 13 and 18), b e muscular ventricular septal defect<br />

(Figs. 14 and 15): i. muscular inlet, ii. muscular outlet (Fig. 14),<br />

iii. trabecular defect (Fig. 15), c e doubly committed ventricular<br />

septal defect (Figs. 16 and 19), d e inlet ventricular septal<br />

defect (Fig. 17).<br />

4.3. Size of ventricular septal defect<br />

The judgment of size of defect is generally made on hemodynamic<br />

grounds (degree of left to right shunt, presence of<br />

volume overload, and pulmonary artery pressure).<br />

Comparative size: According to some authors a VSD size is<br />

defined in relation to aortic root size. Small ventricular septal<br />

defect are defined if less than 1/3rd of aortic root diameter, 1/<br />

3rd to 2/3rd of aortic root diameter considered as moderate<br />

sized defect, and the lesions that are approximate to the size<br />

of aortic root are defined as large VSD. The disadvantage of the<br />

method of classifying defects is that some defects which are<br />

anatomically large can be restricted in terms of shunt and<br />

pressure because of reduction in size by tricuspid valve or<br />

aortic valve.<br />

Hemodynamic classification uses the pressure differential<br />

across the left to right ventricle to classify defects. With isolated<br />

defects, when there is equalization of pressure between<br />

Fig. 18 e 2d echocardiography with various view shows the profilation of perimembranous VSD. A: shows the<br />

perimembranous defect with anterior tilt in subcostal coronal view with anterior tilt showing the defect. B: shows the<br />

parasternal short axis view shows the location of the perimembranous defect from 9 o’clock to 11 o’clock position, C1:<br />

shows the location of the perimembranous VSD with slight posterior tilt toward the tricuspid valve in 2d and color flow<br />

mapping (C2).


212<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

pulmonary stenosis. With severe right ventricular outflow<br />

obstruction, if ventricular septal defect is small, one can get<br />

a turbulent jet of right to left shunt with suprasystemic right<br />

ventricle systolic pressure (Fig. 21).<br />

With the use of color flow mapping with careful interrogation,<br />

one should also look for any left ventricle to right<br />

atrial shunt as high velocity of left ventricle to right atrial<br />

jet can be misinterpreted as elevated right ventricle pressure.<br />

This can happen particularly with ventricular septal<br />

defect which are getting smaller by septal leaflet of<br />

tricuspid valve.<br />

4.5. Direction of shunt<br />

Fig. 19 e 2d echocardiography with parasternal long axis<br />

view shows the doubly committed VSD getting restricted<br />

with right coronary cusp of the aortic valve with significant<br />

prolapse of the right coronary cusp. Though such prolapse<br />

may significantly decrease the left to right shunting and<br />

hence volume overload, it by itself because of the<br />

significant prolapse is an indication of surgery to prevent<br />

the damage to the aortic valve.<br />

two ventricles in absence of pulmonary stenosis, then it is<br />

called as large or nonrestrictive defect. Since right and left<br />

ventricles do not contract exactly simultaneously, there is<br />

always some inequality in the ventricular pressures as estimated<br />

by Doppler technique since it measures instantaneous<br />

pressure difference. A restrictive defect is one in which right<br />

ventricular and pulmonary artery pressures are lower than<br />

left ventricle with pressure gradient of more than 60 mmHg<br />

(ventricular septal defect peak velocity more than 4 m/s), and<br />

moderately restrictive ventricular septal defect with pressure<br />

difference of 25e60 mmHg (ventricular septal defect peak<br />

velocity 2.5e4 m/s).<br />

The VSD may be associated with anterior or posterior<br />

malalignment and these VSDs in the region of the perimembranous<br />

area are generally large VSDs. Those with anterior<br />

malalignment may be associated with narrowing of the<br />

anterior outflow tract (generally pulmonary) as in Tetralogy of<br />

Fallot. Those VSDs associated with posterior malalignment<br />

are associated with narrowing of the posterior outflow<br />

(generally aorta) (Fig. 20 AeC).<br />

4.4. Patterns of shunting across VSD<br />

Nonrestrictive ventricular septal defect with high pulmonary<br />

vascular resistance, direction of flow can be bidirectional<br />

or dominantly right to left depending upon the<br />

severity of pulmonary vascular obstructive disease.<br />

With associated pulmonary stenosis, there may be isolated<br />

right to left shunt depending upon the severity of<br />

With isolated uncomplicated nonrestrictive VSD, pressure<br />

between the two ventricles is similar.<br />

In patients with low pulmonary vascular resistance, dominant<br />

shunt occurs from left to right during systole, and with<br />

increase in left ventricle end diastolic pressure left to right<br />

shunt will persist during diastole also.<br />

A typical ‘M’ shaped flow pattern is being described in<br />

patients with nonrestrictive VSD, explanation for which ise<br />

as left ventricle contraction starts early and last longer than<br />

right ventricle, so with onset of systole flow occur from left<br />

to right, with decrease in degree of shunt during mid systole<br />

as pressure between two ventricles equalized, and in later<br />

part of systole as right ventricle relaxes left to right shunt<br />

dominates.<br />

With restrictive VSD, left to right shunting occurs<br />

throughout systole. In some small muscular VSD, left to<br />

right shunt occurs only during a portion of systole, presumably<br />

because of closure of muscular ventricular septal<br />

defect in mid systole with ventricular contraction. Bidirectional<br />

or right to left shunting can occur with restrictive and<br />

nonrestrictive VSD as described earlier.<br />

4.6. Continuous and pulsed wave Doppler examination<br />

Pulsed and continuous wave Doppler examination is used to<br />

assess 14e16 :<br />

- Direction of shunt across VSD.<br />

- Pressure gradient across the defect (difference of left ventricleeright<br />

ventricle systolic pressure).<br />

- Right ventricle pressure (by VSD gradient and peak gradient<br />

of tricuspid regurgitation jet).<br />

- Diastolic function of both ventricles.<br />

4.7. Pressure gradient across ventricular septal defect<br />

(Fig. 3b)<br />

While taking continuous wave Doppler across VSD, the<br />

cursor should be well aligned with VSD jet on color flow<br />

mapping.<br />

The velocity of the VSD shunt can be determined using the<br />

Bernoulli’s equation. This will give the difference between<br />

the left and right ventricular systolic pressure.<br />

The left ventricular systolic pressure is derived from the<br />

systolic blood pressure (provided there is no left ventricular


indian heart journal 65 (<strong>2013</strong>) 201e218 213<br />

Fig. 20 e A and B shows a large malaligned ventricular septal defect with anterior malalignment of the septum leading to<br />

associated pulmonary stenosis profiled in parasternal long axis view (A) and subcostal view (B). C shows the large<br />

malaligned VSD with anterior malalignment leading to subaortic narrowing profiled in parasternal long axis view.<br />

out flow obstruction), which should be recorded at the time<br />

of Doppler study using appropriate sized BP cuffs.<br />

Right ventricular pressure ¼ Systolic blood pressure VSD jet<br />

peak gradient<br />

This equation has been found to have good correlation<br />

with cardiac catheterization derived right ventricle systolic<br />

pressure.<br />

However, sometimes the jet velocity may not reflect the<br />

interventricular pressure gradient accurately because<br />

proper alignment of the Doppler beam with the jet is not<br />

possible, and that if the defect has some length to it, the<br />

viscous frictional forces make the application of the modified<br />

Bernoulli’s equation inappropriate.<br />

Determining the right ventricular pressure from tricuspid<br />

insufficiency jet velocity (which may be found in some<br />

cases) is also very useful.<br />

4.8. M mode echocardiography (Fig. 22)<br />

Assess the left atrial and left ventricular size to quantitate<br />

shunt across ventricular septal defect.<br />

Right ventricular size and wall thickness, which will reflect,<br />

elevated right ventricular systolic pressure.<br />

For direction of shunt, this is rarely used in daily practice but<br />

depicts best the direction of shunting during the various<br />

phases of a cardiac cycle.<br />

4.9. Interrogation of the atrioventricular and semilunar<br />

valves for regurgitation and stenosis<br />

The tricuspid regurgitation velocity should always be obtained<br />

to predict the right ventricular systolic pressure and<br />

hence indirectly the pulmonary artery pressure if there is no<br />

pulmonary stenosis.<br />

Presence of pulmonary stenosis, and aortic regurgitation<br />

should be evaluated.<br />

Fig. 21 e 2d echocardiography with parasternal long axis<br />

view of a case of doubly committed VSD in a 25-year-old<br />

man with Eisenmenger syndrome showing significant<br />

right to left shunt. The LV dimensions were normal in the<br />

present case with Z score of 1.1.<br />

Fig. 22 e M mode echocardiographic evaluation of a case of<br />

VSD taken in parasternal long axis view. The LV end<br />

diastolic dimensions (LVIDd) is compared to expected for<br />

the weight of the child and a dilated LV with Z score of<br />

more than 2 signifies significant left to right shunt (>2:1)<br />

and hence an indication for intervention.


214<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

The velocities of both atrioventricular valves and semilunar<br />

valves should be taken to rule out any associated abnormality.<br />

The mitral valve gradients may get exaggerated in<br />

the presence of VSD (Fig. 23).<br />

4.10. Pulmonary blood flow to systemic blood flow ratio<br />

As regards quantifying pulmonary and systemic shunt flow<br />

using Doppler echocardiography several methods currently<br />

exists, although none is widely used due to variable<br />

results.<br />

4.11. Assessment of suitability for device closure<br />

Percutaneous closure for mid muscular VSD and perimembranous<br />

VSD can be done. While assessing the child for device<br />

closure of muscular defect, the defect should be at least 5 mm<br />

away from atrioventricular valves and semilunar valves and<br />

there should not be associated other defects requiring cardiopulmonary<br />

bypass. The softer variety of ADOII series can<br />

be used to close the defects in perimembranous region<br />

(particularly in a defect with good aneurysm) and also in<br />

muscular VSD.<br />

4.12. Utility of transesophageal echocardiography<br />

Transesophageal echocardiography has helped in better<br />

visualization of VSD, especially when transthoracic window<br />

is poor, like in adolescents and adults. Straddling and<br />

overriding of the atrioventricular defect can be better<br />

detected by transesophageal echocardiography.<br />

5. Patent ductus arteriosus (PDA) (Table 3)<br />

5.1. Objectives of echocardiography<br />

The presence of a duct<br />

Detailed definition of ductus<br />

B Size of the duct<br />

B Type of duct<br />

The hemodynamic significance of a duct<br />

B Direction of shunt<br />

B Pulmonary arterial pressure<br />

B Quantification of shunt<br />

Associated defects<br />

5.2. Method<br />

Various views to define the ductus are as under 17e20 :<br />

1) Ductal View (Fig. 24) e this uses the high parasternal window<br />

just beneath the left clavicle. After obtaining the short<br />

axis cut of the great vessel visualizing the pulmonary artery<br />

bifurcation the transducer is rotated anticlockwise in<br />

gradual motion. At one point the left pulmonary artery goes<br />

away from view and the duct with adjacent descending<br />

aorta opens. This view in neonates and infants also visualizes<br />

the origin of the left subclavian artery. In patients<br />

with associated coarctation the posterior shelf is also well<br />

visualized.<br />

2) Suprasternal view e<br />

a) Suprasternal long axis view e This is the best view for<br />

visualizing the vertical duct arising from the undersurface<br />

of the transverse arch in patients with pulmonary<br />

atresia. The origin of such ductii is well seen<br />

Fig. 23 e 2d echocardiography with color compare showing the well open mitral valve in 2d (A) and turbulence across mitral<br />

valve in B. The turbulence in color flow mapping across the mitral valve is essentially because of the increased flow across<br />

the mitral valve because of the associated nonrestricted VSD extending from the perimembranous to the inlet septum.


indian heart journal 65 (<strong>2013</strong>) 201e218 215<br />

insertion. In the usual duct, this can be accurately<br />

measured in the ductal view or the modified arch view.<br />

b) Size of the ampulla of duct e it can be best measured in the<br />

modified ductal view.<br />

c) The length of the duct that is necessary to determine adequacy<br />

of coil/device/stent placement and also the need<br />

for evaluation for ADOII series of PDA devices. It is again<br />

best determined by the modified ductal view.<br />

In patients with inadequate windows, the size of the duct<br />

can be determined by the narrowest width of the color flow<br />

across the duct. This, however, always overestimates the<br />

ductal size and gives only a rough estimate.<br />

5.4. Hemodynamic significance<br />

Hemodynamic significance of ductus arteriosus can be<br />

assessed by evidence of volume overload of LA and LV, direction<br />

of shunt, and pulmonary arterial pressure.<br />

Fig. 24 e 2d echocardiography with color flow mapping in a<br />

high parasternal view (ductal view) showed the PDA with<br />

left to right shunt across. There is a good ampulla of the<br />

ductus with narrowing of the ductus before its insertion<br />

into the pulmonary artery.<br />

but the insertion point at the pulmonary artery<br />

required further anterior tilt. This is because of the<br />

tortuous nature of such ductii. In patients with<br />

discordant ventriculo-arterial connection (e.g. transposition<br />

of great vessels), the duct can be visualized<br />

very well in its entire length in this view.<br />

b) Suprasternal short axis view e this is the classical<br />

short axis arch view and can visualize those rare ductii<br />

which arises form the base of the left subclavian artery<br />

and descends straight down to insert into the left<br />

pulmonary artery. If aortic arch is right sided and the<br />

patient has pulmonary stenosis physiology. The entire<br />

length of the duct can be seen in one view because<br />

unlike in those patients with vertical duct it does not<br />

follow a tortuous course.<br />

c) Modified ductal view e this a less well described view<br />

to visualize the usual duct. It has the advantage of<br />

visualizing the duct in its entire length and most<br />

closely mimics the lateral angiogram performed during<br />

cardiac catheterization. From the usual suprasternal<br />

long axis view the transducer is rotated<br />

anticlockwise. A slight anterior tilt then shows the<br />

duct from its ampullary part to its insertion and accurate<br />

measurements can be made.<br />

5.3. Measurements of the duct<br />

Various measurements on the duct by echocardiography<br />

include:<br />

a) Size of the narrowest part of the duct e in the majority of<br />

cases this would be at the site of pulmonary artery<br />

Chamber dimensions:<br />

Left atrial enlargement signifies increased pulmonary<br />

venous return because of left-to-right ductal shunting.<br />

The reference measure is the ratio of the left atria to aorta<br />

at the level of the aortic valve (the LA: Ao ratio) by M mode<br />

echocardiography in parasternal long axis view.<br />

The aortic root does not enlarge significantly with even<br />

extremely large patent ductus arteriosus.<br />

A LA: Ao ratio >1.3:1 indicates a significant shunt.<br />

LV will enlarge as cardiac output increases with both<br />

increased pulmonary venous return and with increased<br />

diastolic run-off from the systemic circulation.<br />

5.5. Direction of shunt and pulmonary arterial pressure<br />

On color flow mapping, small duct with normal pulmonary<br />

artery pressure is displayed as a mosaic flow from<br />

descending aorta to pulmonary artery. With large duct, and<br />

low pulmonary vascular resistance, the duct jet appears as<br />

predominantly red flow with minimal aliasing.<br />

In patients with severe pulmonary arterial hypertension, on<br />

color flow mapping, there will be bidirectional shunt.<br />

With suprasystemic pulmonary artery pressure as in<br />

obstructed total anomalous pulmonary venous connection<br />

a restrictive duct will show turbulent high velocity right to<br />

left flow in systole and diastole in the descending aorta. This<br />

can give signals very similar to coarctation of aorta.<br />

5.6. Continuous wave Doppler examination of ductus<br />

arteriosus (Fig. 3A)<br />

By the use of continuous wave Doppler, direction of shunt in<br />

relation to cardiac cycle and pulmonary arterial pressure<br />

(systolic blood pressure minus pressure gradient cross<br />

duct ¼ systolic pulmonary arterial pressure) can be detected<br />

accurately.<br />

With isolated left to right shunt, with small to moderate<br />

sized patent ductus arteriosus and normal or mildly<br />

elevated pulmonary artery pressure, Doppler examination


216<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

of duct shows continuous flow toward the transducer with<br />

peak in late systole.<br />

In large duct with pulmonary arterial hypertension, there<br />

will be bi-directional shunting on Doppler imaging of duct,<br />

right to left in systole and left to right in diastole.<br />

With increasing pulmonary vascular resistance as with no<br />

step up in oxygen saturation above and below the duct, peak<br />

of right to left shunt appear early in systole.<br />

With further rise in pulmonary vascular resistance, right to<br />

left shunt begins in diastole extending to systole and right to<br />

left shunt if present occurs only in late systole to early<br />

diastole.<br />

With duct dependent systemic circulation and severe pulmonary<br />

arterial hypertension, there will be isolated right to<br />

left shunting across the duct.<br />

5.7. Limitations of echocardiographic imaging of the<br />

duct<br />

There are several limitations of profilation of duct by twodimensional<br />

imaging<br />

1) The primary limitation of echocardiography is the restriction<br />

imposed by limited acoustic windows. The duct is<br />

profiled in the direction of lateral resolution of the transducer,<br />

so it is difficult to visualize with certainty very small<br />

duct in small babies. High frequency probe with excellent<br />

lateral resolution is needed.<br />

2) If the duct is long and tortuous, it may be difficult to profile<br />

whole length of the duct<br />

3) Poor acoustic windows as in adults with thick chest.<br />

6. Aortopulmonary window (Fig. 25)<br />

Aortopulmonary window or aortopulmonary septal defect<br />

accounts for 0.2e0.6% of patients with congenital heart<br />

defects. Nearly half of all patients have associated cardiac<br />

lesions, including aortic origin of the right pulmonary artery,<br />

type A interruption of the aortic arch, Tetralogy of Fallot, and<br />

anomalous origin of the right or left coronary artery from the<br />

pulmonary artery and right aortic arch. More rarely, it is<br />

associated with ventricular septal defect, pulmonary or aortic<br />

atresia, D-transposition, and tricuspid atresia<br />

Objectives of echocardiography<br />

Diagnosis<br />

Type of aortopulmonary window<br />

Associated heart defects<br />

Operability<br />

Two-dimensional echocardiography usually can accurately<br />

diagnose the aortopulmonary septal defect.<br />

Views, which are most useful for diagnosis, are parasternal<br />

short axis at the level of great vessels, subcostal coronal<br />

view of left ventricular outflow tract and the suprasternal<br />

views.<br />

In all these views, the wall separating aorta and pulmonary<br />

artery is aligned in the direction of lateral resolution, so<br />

great care is needed to differentiate true defect from artifactual<br />

dropout. A ‘T’ artifact at the edges of the defect will<br />

distinguish it from normal dropout.<br />

Color flow mapping: Color flow mapping is used to<br />

demonstrate flow through the defect.<br />

With large defect, which is usually the case, the flow appear<br />

laminar, low velocity, bidirectional flow across the defect.<br />

Smaller defect, a continuous high velocity left to right jet is<br />

usually present.<br />

With low pulmonary vascular resistance, evidence of aortic<br />

run-off can be detected in ascending and descending aorta<br />

in contrast to patent ductus arteriosus.<br />

Fig. 25 e 2d echocardiography with color comparison in parasternal short axis view showing a large AP widow in 2d (A)and<br />

color Doppler (B).


indian heart journal 65 (<strong>2013</strong>) 201e218 217<br />

Fig. 26 e 2d echocardiography with color mapping comparison showing the Gerbode defect with left to right shunt.<br />

7. Gerbode defect (Figs. 26 and 27)<br />

The lesions such as Gerbode defect suggest LV to RA shunt and<br />

essentially lead to volume overload of the RA and the RV.<br />

In such cases the RA dimensions and features of right atrial<br />

volume overload need to be looked at. The features are<br />

essentially same as that of the atrial septal defect.<br />

The point to remember is that the pressure difference across<br />

tricuspid valve taken in these circumstances may be<br />

fallacious as one may pick up the left ventricle to RA<br />

gradient which will be systemic (mistaking it to be that of<br />

the tricuspid velocity signal).<br />

The shunt lesions such as pulmonary AV fistulas and<br />

coronary AV fistulas and systemic AV fistulas are not specifically<br />

covered in the present section. For the purpose of<br />

completion, they will represent the left to right shunt and<br />

would lead to the chamber enlargement of the chamber into<br />

which they drain.<br />

Fig. 27 e 2d echocardiography with color flow comparison in parasternal short axis view shows the Gerbode defect with left<br />

to right shunt.


218<br />

indian heart journal 65 (<strong>2013</strong>) 201e218<br />

Conflicts of interest<br />

All authors have none to declare.<br />

references<br />

1. Awasthy N, Radhakrishnan S, Shrivastava S. Double orifice<br />

mitral valve with PDA. J Indi Acad Echo. <strong>2013</strong>;24:48e50.<br />

2. Awasthy N, <strong>Mar</strong>wah A, Sharma R, Dalvi B. Anomalous origin<br />

of the left coronary artery from the pulmonary artery with<br />

patent ductusarteriosus: a must to recognize entity. Eur J<br />

Echocardiogr. 2010;11:E31.<br />

3. Awasthy N, <strong>Mar</strong>wah A, Sharma R. Occult anomalous<br />

origin of the left coronary artery from the pulmonary<br />

artery with ventricular septal defect. Ann Pediatr Cardiol.<br />

2011;4:62e64.<br />

4. Awasthy N, Ambadkar P, Radhakrishnan S, Iyer KS.<br />

Lutembacher syndrome with unroofed left superior vena<br />

cava: a diagnostic dilemma. Pediatr Cardiol. 2012;24:1e2.<br />

5. Awasthy N, Tomar M, Radhakrishnan S, Iyer KS. Constriction<br />

juxta-ductal aorta rapid progression obstruction a newborn.<br />

Ann Pediatr Cardiol. 2010;3(2):181e183.<br />

6. Shrivastava S, Radhakrishnan R, Tomar M, eds.<br />

Echocardiography in Pediatric <strong>Heart</strong> Disease. 1st ed. Siddharth<br />

Publications; 2009.<br />

7. Vermilion PR. Basic physical principles. In: Snider AR,<br />

Serwer AG, Ritter SB, eds. Echocardiography in Pediatric <strong>Heart</strong><br />

Disease. 2nd ed. Mosby; 1997:1e10.<br />

8. Dillon JC, Wayman AE, Feigenbaum H, Eggleton RC,<br />

Johnston K. Cross-sectional echocardiographic<br />

examination of the interatrial septum. Circulation.<br />

1977;55:115e120.<br />

9. Child JS. Echo-Doppler and color-flow imaging in congenital<br />

heart disease. Cardiol Cln. 1990;8:289e313.<br />

10. Nasser FN, Tajik AJ, Seword JB, Hagler DJ. Diagnosis of sinus<br />

venosus atrial septal defect by two-dimensional<br />

echocardiography. Mayo Clin Proc. 1981;56:568e572.<br />

11. Assessment of right to left shunt flow in atrial septal defect by<br />

transesophageal color and pulsed Doppler echo cardiography.<br />

J Am Soc Echocardiogr. 1994;7(5):506e515.<br />

12. Bierman FZ, Fellows K, Williams RG. Prospective identification<br />

of ventricular septal defect in infancy using subxiphoid twodimensional<br />

echocardiography. Circulation. 1980;6:807e817.<br />

13. Capelli H, Andrads JL, Somerville J. Classification of the site of<br />

ventricular septal defect by two-dimensional<br />

echocardiography. Am J Cardiol. 1983;51:1474e1480.<br />

14. Ortiz E, Robinson PJ, Deanfield JE, Franklin R, Macartney FJ,<br />

Wyse RK. Localization of ventricular septal defects by<br />

simultaneous display of superimposed colour Doppler and<br />

cross sectional echocardiographic images. Br <strong>Heart</strong> J.<br />

1985;54:53e60.<br />

15. Helmcke F, de Souza A, Nanda NC, et al. Two-dimensional<br />

and color Doppler assessment of ventricular septal defect of<br />

congenital origin. Am J Cardiol. 1989;63:1112e1116.<br />

16. Murphy DJ, Ludomirsky A, Huhta JC. Continuous wave<br />

Doppler in children with ventricular septal defect:<br />

noninvasive estimation of interventricular pressure gradient.<br />

Am J Cardiol. 1986;57:428e432.<br />

17. Huhta JC, Cohen M, Gutgesell HP. Patency of the ductus<br />

arteriosus in normal neonates: two dimensional<br />

echocardiography vs Doppler assessment. J Am Coll Cardiol.<br />

1984;4:561e564.<br />

18. Silverman NH. Patent ductus arteriosus. In: Pediatric<br />

Echocardiography. Baltimore, MD: Williams & Wilkins;<br />

1993:167e177.<br />

19. Liao PK, Su WJ, Hung JS. Doppler echocardiographic flow<br />

characteristic of isolated patent ductus arteriosus: better<br />

delineation by Doppler color flow mapping. J Am Coll Cardiol.<br />

1988;12:1285e1291.<br />

20. Swensson RE, et al. Real time Doppler color flow mapping for<br />

detection of patent ductus arteriosus. Pediatr Cardiol.<br />

1986;8:1105.


222<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

patients without baseline “SCD-HeFT criteria” (left ventricular<br />

ejection fraction >0.35 or NYHA class I), 125 were<br />

evaluated after 5.5 2 months. Of these 227 patients, 13<br />

(10%) developed “SCD-HeFT criteria” (group B1), 111 (89%)<br />

remained without “SCD-HeFT criteria” (group B2), and 1<br />

(1%) had worsened to NYHA class IV. The 10-year mortality/heart<br />

transplantation and sudden death/sustained<br />

ventricular arrhythmia rate was 57% and 37% in group A1,<br />

23% and 20% in group A2 (p < 0.001 for mortality/heart<br />

transplantation and p e 0.014 for sudden death/sustained<br />

ventricular arrhythmia vs. group A1), 45% and 41% in group<br />

B1 ( p e NS vs. group A1), 16% and 14% in group B2 ( p e NS<br />

vs. group A2), respectively.<br />

Conclusion: Two thirds of patients with idiopathic<br />

dilated cardiomyopathy and “SCD-HeFT criteria” at presentation<br />

did not maintain implantable cardioverterdefibrillator<br />

indications 3e9 months later with optimal<br />

medical therapy. Their long-term outcome was excellent,<br />

similar to that observed for patients who had never met the<br />

“SCD-HeFT criteria.”<br />

1. Perspective<br />

Since the publication of the SCD-HeFT and the DEFINITE<br />

trials, treatment with ICD for the primary prevention of<br />

sudden cardiac death (SCD) has been extended to patients<br />

with idiopathic dilated cardiomyopathy (IDC), who have a<br />

LVEF of 0.35 and who are classified as NYHA II or III (“SCD-<br />

HeFT criteria,” class I B indication). The appropriate timing<br />

for ICD implantation, however, is still uncertain. Current<br />

guidelines suggest that an ICD should be considered in<br />

addition to medical therapy, but many patients are treated<br />

with an ICD without evidence-based indications, mainly<br />

because of newly diagnosed heart failure and before treatment<br />

optimization. This study evaluated the proportion of<br />

patients with and without potential indications for ICD<br />

implantation at presentation and the long-term prognosis of<br />

patients with initial ICD indications but who improved after<br />

optimization of medical treatment. It also compared the<br />

long-term outcome of “improved” patients to those maintaining<br />

“SCD-HeFT criteria” and those who never met “SCD-<br />

HeFT criteria.”<br />

This trial included only patients who were not on beta<br />

blockers. After initial assessment, optimization of medical<br />

treatment was achieved with gradually up-titrating doses of<br />

beta blockers and ACEI/ARB at the highest tolerated dose over<br />

a period of 3e9 months.<br />

The main results of the present study are: 1) 50% patients<br />

had SCD-HeFT criteria at first assessment and would have<br />

otherwise received an ICD. 2) 2/3rd of patients with SCD-HeFT<br />

criteria at baseline “improved” and no longer maintained SCD-<br />

HeFT criteria 5.5 months after starting beta blocker and ACEI<br />

treatment. 3) The long-term SCD were similar in “improved”<br />

patients and in those without SCD-HeFT criteria, suggesting<br />

ICD implantation should not be done in most patients with<br />

low LVEF and HF symptoms before optimization of medical<br />

treatment. 4) SCD (4 patients e 2%) was similar in patients<br />

both with and without SCD-HeFT criteria before second<br />

evaluation at 3e9 months, confirming the difficulty of stratifying<br />

risk of SCD at first evaluation.<br />

This study emphasizes how important is the optimization<br />

of medical therapy in patients initially presenting with ICD<br />

indications and ICD implantation can be avoided in the<br />

majority of such patients. Even in USA, nearly 22.5% of<br />

patients with an ICD did not meet the evidence-based criteria<br />

for implantation, mainly because of newly diagnosed<br />

HF (62%). Such unnecessary ICD implantations should be<br />

avoided because of economic issues (especially in a developing<br />

country like India), the risk of complications associated<br />

with implantation and inappropriate shocks (in w25%<br />

of patients).<br />

What then is the waiting period for ICD implantation after<br />

onset of HF symptoms in patients with LVEF 35%? Well, we<br />

have no clear-cut answer. Data from DEFINITE trial suggest<br />

early ICD implantation (


indian heart journal 65 (<strong>2013</strong>) 219e228 223<br />

patients who were on dialysis. This study compared Cinacalcet<br />

with placebo in 3883 patients undergoing dialysis. Limited<br />

number of interventions are found to improve cardiovascular<br />

health in CKD undergoing dialysis. Secondary hyperparathyroidism<br />

has emerged as one of the most important<br />

risk factors for cardiovascular disease and thus, high rate of<br />

death and cardiovascular events in end stage renal disease. 3<br />

Although this trial result is non-definitive but after adjustment<br />

to the baseline characteristics there was nominally significant<br />

12% reduction in cardiovascular risk, 15% reduction in<br />

primary composite end point and 17% reduction in mortality.<br />

One has to understand the context of the patients who are<br />

on dialysis, frequently have poor health and high mortality<br />

(20% in United States) and morbidity (median 2 hospitalization<br />

and 12 hospital days per year). Patients have multi organ<br />

involvement with average pill burden of 19. Cinacalcet<br />

reduces parathyroidectomy by 50%. The study includes<br />

Cohorts from various geographic area, race, ethnicity, age and<br />

underlying kidney and cardiovascular diseases.<br />

Analysis adjusted for baseline characteristics on taking<br />

into account the effect of parathyroidectomy and kidney<br />

transplantation a nominally significant reduction on death on<br />

first Myocardial Infarction, hospitalization for unstable<br />

angina, heart failure and peripheral vascular disease (risk<br />

reduction 10e15% and absolute reduction of 2e3%).<br />

references<br />

1. Kidney disease improving global outcomes (KDIGO) CKD-MBD<br />

Work Group. Kidney Int Suppl. 2009;76:S1eS130.<br />

2. Cunningham J, et al. Kidney Int. 2005;68:1793.<br />

3. Chertow GM, et al. Am Soc Nephrol. 2007;2:898e905.<br />

Ajay Kumar Sinha<br />

Associate Editor, IHJ, India<br />

E-mail address: sinha_ajaykr@yahoo.co.in<br />

Gregg W. Stone, Alexandre Abizaid, Sigmund Silber, et al.,<br />

Prospective, randomized, multicenter evaluation of a polyethylene<br />

terephthalate micronet mesh-covered stent<br />

(MGuard) in ST-segment elevation myocardial infarction: the<br />

MASTER trial. J Am Coll Cardiol 60 (2012) 1975e1984<br />

1. Introduction<br />

The enemy of revascularization using primary PCI during<br />

STEMI intervention is risk of distal embolization with capillary<br />

plugging which leads to reduced tissue reperfusion. Several<br />

pharmacological agents, as well as mechanical devices (i.e.<br />

manual aspiration catheters/mechanical thrombectomy,<br />

proximal and distal protection devices) were introduced, in<br />

the last years, to reduce the risk of angiographic complications<br />

during percutaneous coronary intervention and to<br />

improve myocardial reperfusion. 1,2 Despite the use of these<br />

agents still distal embolization is common which leads to noreflow<br />

phenomenon. 1,2<br />

Recently, the MGuard stent (InspireMD, Tel Aviv, Israel), a<br />

bare metal stent covered by micron level mesh, which allows<br />

to prevent distal embolization by blocking the athero-thrombi<br />

prolapse through the stent struts during deployment. 3<br />

Dr. Gregg W. Stone presented the late-breaking findings<br />

from the MASTER study were presented in a late-breaking<br />

session at TCT 2012 that revealed a new stent in STEMI<br />

patients undergoing emergent PCI to increase the rate of complete<br />

ST-segment resolution compared with conventional BMS<br />

and DES.<br />

2. Objectives<br />

MASTER study sought to evaluate the potential utility of a<br />

novel polyethylene terephthalate micronet mesh-covered<br />

stent (MGuard) in patients with acute ST-segment elevation<br />

myocardial infarction (STEMI) undergoing percutaneous coronary<br />

intervention (PCI).<br />

3. Study design<br />

The MASTER trial is a prospective, multicenter, randomized<br />

study designed to compare the incidence of complete<br />

(70%) ST-segment resolution with PCI using bare metal or<br />

drug-eluting stents (the control arm) versus PCI with the<br />

MGuard stent, measured 60e90 min after the last angiogram<br />

(primary endpoint).<br />

Secondary endpoints include the rates of TIMI flow and<br />

myocardial blush, and clinical outcomes through 1-year<br />

follow-up.<br />

The study has enrolled 432 patients with STEMI undergoing<br />

primary or rescue angioplasty within 12 h of symptom<br />

onset, and includes sub studies with cardiac magnetic resonance<br />

imaging and quantitative coronary angiography to<br />

evaluate infarct size, micro vascular obstruction and<br />

angiographic restenosis.<br />

Study population for trial enrolled a total 433 STEMI patients<br />

who presented within 12 h of symptom onset, at 50 sites in 9<br />

countries.<br />

These enrolees were randomly assigned to receive commercially<br />

available stents (n ¼ 216; 60% BMS; 40% DES) or<br />

MGuard (n ¼ 217). The two groups had equal baseline<br />

characteristics. 3<br />

4. Results<br />

Significantly more patients treated with the MGuard<br />

EPS achieved the primary endpoint of post-procedure<br />

of complete ST resolution (a measure of blood flow<br />

restoration to the heart muscle) compared to control arm<br />

(57.8% vs. 44.7%, p ¼ 0.008), a relative improvement of 29%<br />

[Fig. 2].<br />

When compared to control, the MGuard EPS showed a significant<br />

improvement in coronary artery blood flow,<br />

including (1): superior rates of restoring normal blood flow<br />

(TIMI 3 flow) (91.7% vs. 82.9%, p ¼ 0.006, a relative<br />

improvement of 10.6%); and (2) significantly less incomplete<br />

blood flow (TIMI 0/1 flow) post PCI (1.8% vs. 5.6%, p ¼ 0.01, a<br />

relative improvement of 67.9%).<br />

The trial showed a trend toward lower mortality (0% vs.<br />

1.9%, p ¼ 0.06) at 30 days and smaller infarct size as


indian heart journal 65 (<strong>2013</strong>) 219e228 223<br />

patients who were on dialysis. This study compared Cinacalcet<br />

with placebo in 3883 patients undergoing dialysis. Limited<br />

number of interventions are found to improve cardiovascular<br />

health in CKD undergoing dialysis. Secondary hyperparathyroidism<br />

has emerged as one of the most important<br />

risk factors for cardiovascular disease and thus, high rate of<br />

death and cardiovascular events in end stage renal disease. 3<br />

Although this trial result is non-definitive but after adjustment<br />

to the baseline characteristics there was nominally significant<br />

12% reduction in cardiovascular risk, 15% reduction in<br />

primary composite end point and 17% reduction in mortality.<br />

One has to understand the context of the patients who are<br />

on dialysis, frequently have poor health and high mortality<br />

(20% in United States) and morbidity (median 2 hospitalization<br />

and 12 hospital days per year). Patients have multi organ<br />

involvement with average pill burden of 19. Cinacalcet<br />

reduces parathyroidectomy by 50%. The study includes<br />

Cohorts from various geographic area, race, ethnicity, age and<br />

underlying kidney and cardiovascular diseases.<br />

Analysis adjusted for baseline characteristics on taking<br />

into account the effect of parathyroidectomy and kidney<br />

transplantation a nominally significant reduction on death on<br />

first Myocardial Infarction, hospitalization for unstable<br />

angina, heart failure and peripheral vascular disease (risk<br />

reduction 10e15% and absolute reduction of 2e3%).<br />

references<br />

1. Kidney disease improving global outcomes (KDIGO) CKD-MBD<br />

Work Group. Kidney Int Suppl. 2009;76:S1eS130.<br />

2. Cunningham J, et al. Kidney Int. 2005;68:1793.<br />

3. Chertow GM, et al. Am Soc Nephrol. 2007;2:898e905.<br />

Ajay Kumar Sinha<br />

Associate Editor, IHJ, India<br />

E-mail address: sinha_ajaykr@yahoo.co.in<br />

Gregg W. Stone, Alexandre Abizaid, Sigmund Silber, et al.,<br />

Prospective, randomized, multicenter evaluation of a polyethylene<br />

terephthalate micronet mesh-covered stent<br />

(MGuard) in ST-segment elevation myocardial infarction: the<br />

MASTER trial. J Am Coll Cardiol 60 (2012) 1975e1984<br />

1. Introduction<br />

The enemy of revascularization using primary PCI during<br />

STEMI intervention is risk of distal embolization with capillary<br />

plugging which leads to reduced tissue reperfusion. Several<br />

pharmacological agents, as well as mechanical devices (i.e.<br />

manual aspiration catheters/mechanical thrombectomy,<br />

proximal and distal protection devices) were introduced, in<br />

the last years, to reduce the risk of angiographic complications<br />

during percutaneous coronary intervention and to<br />

improve myocardial reperfusion. 1,2 Despite the use of these<br />

agents still distal embolization is common which leads to noreflow<br />

phenomenon. 1,2<br />

Recently, the MGuard stent (InspireMD, Tel Aviv, Israel), a<br />

bare metal stent covered by micron level mesh, which allows<br />

to prevent distal embolization by blocking the athero-thrombi<br />

prolapse through the stent struts during deployment. 3<br />

Dr. Gregg W. Stone presented the late-breaking findings<br />

from the MASTER study were presented in a late-breaking<br />

session at TCT 2012 that revealed a new stent in STEMI<br />

patients undergoing emergent PCI to increase the rate of complete<br />

ST-segment resolution compared with conventional BMS<br />

and DES.<br />

2. Objectives<br />

MASTER study sought to evaluate the potential utility of a<br />

novel polyethylene terephthalate micronet mesh-covered<br />

stent (MGuard) in patients with acute ST-segment elevation<br />

myocardial infarction (STEMI) undergoing percutaneous coronary<br />

intervention (PCI).<br />

3. Study design<br />

The MASTER trial is a prospective, multicenter, randomized<br />

study designed to compare the incidence of complete<br />

(70%) ST-segment resolution with PCI using bare metal or<br />

drug-eluting stents (the control arm) versus PCI with the<br />

MGuard stent, measured 60e90 min after the last angiogram<br />

(primary endpoint).<br />

Secondary endpoints include the rates of TIMI flow and<br />

myocardial blush, and clinical outcomes through 1-year<br />

follow-up.<br />

The study has enrolled 432 patients with STEMI undergoing<br />

primary or rescue angioplasty within 12 h of symptom<br />

onset, and includes sub studies with cardiac magnetic resonance<br />

imaging and quantitative coronary angiography to<br />

evaluate infarct size, micro vascular obstruction and<br />

angiographic restenosis.<br />

Study population for trial enrolled a total 433 STEMI patients<br />

who presented within 12 h of symptom onset, at 50 sites in 9<br />

countries.<br />

These enrolees were randomly assigned to receive commercially<br />

available stents (n ¼ 216; 60% BMS; 40% DES) or<br />

MGuard (n ¼ 217). The two groups had equal baseline<br />

characteristics. 3<br />

4. Results<br />

Significantly more patients treated with the MGuard<br />

EPS achieved the primary endpoint of post-procedure<br />

of complete ST resolution (a measure of blood flow<br />

restoration to the heart muscle) compared to control arm<br />

(57.8% vs. 44.7%, p ¼ 0.008), a relative improvement of 29%<br />

[Fig. 2].<br />

When compared to control, the MGuard EPS showed a significant<br />

improvement in coronary artery blood flow,<br />

including (1): superior rates of restoring normal blood flow<br />

(TIMI 3 flow) (91.7% vs. 82.9%, p ¼ 0.006, a relative<br />

improvement of 10.6%); and (2) significantly less incomplete<br />

blood flow (TIMI 0/1 flow) post PCI (1.8% vs. 5.6%, p ¼ 0.01, a<br />

relative improvement of 67.9%).<br />

The trial showed a trend toward lower mortality (0% vs.<br />

1.9%, p ¼ 0.06) at 30 days and smaller infarct size as


224<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

Fig. 1 e AeD The MGuard stent has a 316L stainless steel frame with 100-mm strut thickness. It is manufactured in<br />

diameters ranging from 2.0 to 4.0 mm and in lengths ranging from 11 to 39 mm. The crossing profile ranges from 1.0 to<br />

1.3 mm. The MGuard prime stent is the MGuard prime stent is similar in configuration, but has an L605 cobalt chromium<br />

alloy frame with 80-mm strut thickness and slightly lower crossing profile. It is manufactured in diameters ranging from 2.5<br />

to 4.0 mm and in lengths ranging from 13 to 38 mm. The polyethylene terephthalate micronet is identical on both stents and<br />

has a fiber width of 20 mm and an expanded aperture size of 150 3 180 mm.<br />

measured by post-procedure cardiac MRI (17.1 g vs. 22.3 g,<br />

p ¼ 0.27) in the MGuard EPS arm versus control.<br />

There was no difference between the groups in the secondary<br />

endpoint of myocardial blush grade (MBG), which is an<br />

angiographic measure of blood flow to the cardiac muscle<br />

(MBG2/3 83.9% vs. 84.7%, p ¼ 0.81). 3<br />

5. Conclusions<br />

Among patients with acute STEMI undergoing emergent<br />

PCI, the MGuard micronet mesh-covered stent com e pared<br />

with conventional metal stents resulted in superior rates<br />

of epicardial coronary flow and complete ST-segment<br />

resolution.<br />

6. Clinical perspective<br />

Suboptimal myocardial reperfusion after PCI in STEMI is<br />

common and results in increased infarct size and mortality<br />

(5-year mortality 18.2% with no-reflow vs. patients with good<br />

reflow9.5%). The no-reflow is strong predictor of 5-year mortality[Fig.<br />

3]. 2<br />

Fig. 2 e The primary endpoint of complete ST resolution<br />

was achieved in 29% more compared to routine PCI using<br />

BMS/DES [control arm] while partial and absent ST<br />

resolution comparable to control arm.<br />

Fig. 3 e No-reflow defined as TIMI 0/1/2, or TIMI 3<br />

with MBG 0/1. G. Ndrepepa et al compared 5-year<br />

outcome of 410 patients with no-reflow vs. 996<br />

patients with reflow. (Kaplan-Meier estimates of 5-year<br />

mortality 18.2% and 9.5%, respectively; odds ratio: 2.02;<br />

95% confidence interval: 1.44 to 2.82; p < 0.001).


indian heart journal 65 (<strong>2013</strong>) 219e228 225<br />

Covered stents have been tried for embolic protection<br />

before but they used a tightly woven material that ended up<br />

squeezing friable material out like toothpaste rather than<br />

trapping it. 4 The high density of the covered stent is detrimental<br />

to healing and endothelization process, and also<br />

blocks all side branches large and small.<br />

7. MGuard stent: the technology<br />

InspireMD has developed its proprietary stent system technology,<br />

MGuardä which is CE-<strong>Mar</strong>k approved.<br />

The MGuard is a novel thin-strut metal stent [a balloonexpandable<br />

metallic scaffold] with mesh sleeve fibers of<br />

polyethylene terephthalate micronet attached to its outer<br />

surface designed to trap and exclude thrombus and friable<br />

atheromatous debris to prevent distal embolization<br />

[Fig. 1AeD].<br />

The net is made of a single knitted PET fiber, and it is<br />

attached only to the proximal and distal edges of the stent.<br />

The net expands seamlessly during stent deployment.<br />

MGuard’s net is completely bio-stable. 5<br />

MGuard is deployed exactly like a typical balloon inflated<br />

stent after pre-dilatation if necessary and post dilatation is<br />

recommended whenever there is situation of incomplete<br />

apposition after stent deployment.<br />

The net reduces the risk of plaque rupture and embolization<br />

providing double protection during and post-procedure.<br />

MGuard addresses risks associated with distal embolization<br />

and no-reflow which simplifies primary PCI and SVG interventions<br />

as it is designed for thrombus containing lesions.<br />

Currently, MGuard is not recommended to be used in bifurcation<br />

lesions. 4<br />

8. MGuard stent: evidence<br />

After successful use of MGuard in First in Man trial in 2008,<br />

11 Single-Arm Trials are done with 630 patients with 458<br />

STEMI patients. They evaluated blush grade, ST-segment<br />

resolution and MACE from 30 days to 1 year. Some of the<br />

important trials are summarized in Table 1. The Landmark<br />

trials were the INSPIRE trial 6 which addressed SVG and ACS<br />

Fig. 4 e A showing comparison of blush grade and their<br />

predictor for adverse events for bare metal stent [BMS],<br />

thrombo-aspiration [TA] and MGuard. B showing<br />

comparison of ST resolution and their predictor for adverse<br />

events for bare metal stent [BMS], thrombo-aspiration [TA]<br />

and MGuard. Data on this slide for bare metal stent [BMS],<br />

thrombus aspiration [TA] and MGuard are derived from<br />

different sources. Please note a head to head study has not<br />

been conducted. BMS (n [ 665, TAPAS & EXPORT Trials) TA<br />

(n [ 655, TAPAS & EXPORT trial aspiration arm) MGuard<br />

(n [ 234, MAGICAL, Piscione, iMOS, Lindefjeld, Gaul).<br />

patients using MGUARD stent and MAGICAL trial<br />

7 had<br />

STEMI patients but their sample volume was small. Piscione<br />

et al 9 showed complete ST resolution with 100% device<br />

success rate.<br />

The iMOS registry is single arm prospective ‘real world’<br />

registry which is ongoing registry with target population of<br />

Table 1 e Summery of trials of MGuard stent.<br />

Author/PI Patients Year of publication Highlights<br />

MASTER trial Gregg Stone et al 3 433 2012 RCT, STEMI patients, randomized 1:1 MGuard vs. BMS/DES.<br />

Superior rates of complete ST resolution (57.8% vs. 44.7%,<br />

p ¼ 0.008). Superior rates of TIMI 3 (91.7% vs. 82.9%, p ¼ 0.006),<br />

0% mortality at 30 days<br />

INSPIRE trial A. Abizaid et al 6 30 2010 SVG and native coronaries including ACS, no EPD used, 3.3%<br />

MACE at 30 days, 17% TVR at 1 year 0% death at 1 year<br />

MAGICAL trial D. Dudek et al 7 60 2010 STEMI patients, multicenter, 90% TIMI flow 3, 73% blush grade<br />

3, 0% MACCE at 30 days, 1.7% MACCE at 6 months*<br />

Interim analysis of a real world<br />

registry (iMOS registry)<br />

A. Danzi et al 8 211 2010 77% STEMI, 87% visible thrombus, 96% TIMI 3 flow, 98%<br />

procedural success, 3.8% MACE at 30 days<br />

Federico Piscione et al 9 100 2009 STEMI patients (n ¼ 84, excluding cardiogenic shock) 100%<br />

device success, final cTFC ¼ 17.2, 90% MBG 3 90% complete<br />

ST-segment resolution


226<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

1000 patients. Primary endpoint at 6 months of MACE of<br />

Ischemia related death, myocardial infarction (Q wave and<br />

non-Q wave), target lesion revascularization (PTCA or CABG).<br />

They enrolled 81% of AMI patients with 77% of STEMI.<br />

Thrombus was visible in 87% of the cases and 65% of the<br />

patients showed initial TIMI flow of 0/1. Device success was<br />

98% and 96% of the patients gained TIMI 3 flow postprocedure.<br />

STEMI subgroup clinical analysis revealed a 30<br />

days accumulative MACE of 2.5% with 0% TLR. 8<br />

As shown in Fig. 4 which compares outcomes of MGuard<br />

stent versus bare metal stent and thrombus aspiration trials<br />

which clearly shows benefit of use of MGuard stent in terms of<br />

blush grade and ST-segment resolution. Though this comparison<br />

is not head to head comparison but trials using bare<br />

metal stent and thrombus aspiration were compared with<br />

similar parameters of myocardial blush grade and ST-segment<br />

resolution. This comparison showed 84.6% MBG grade 3<br />

with MGuard as compared to only 44% with use of thrombus<br />

aspiration catheter and >70% ST resolution was seen in 79.6%<br />

compared to 56.6% in patients of trials of thrombus aspiration.<br />

But these data need careful assessment with statistical analysis<br />

and there is further need of RCTs comparing them. 10<br />

Based on these earlier trials Gregg Stone et al conducted<br />

MASTER trial to prove use of novel stenteMGuard and recommended<br />

in any PCI when a stent implantation is needed<br />

and there is a risk of complication due to distal embolization<br />

of plaque or thrombus. 3<br />

Results of the study showed >70% resolution of STsegment<br />

in MGuard 29% relative to control arm of routine PCI<br />

using either BMS or DES in patients undergoing primary PCI<br />

with thrombus containing lesions but partial or absent resolution<br />

of ST-segment was comparable to control arm.<br />

But this study was limited by the fact that the operators<br />

and research coordinators were not blinded to stent assignment,<br />

which possibly introduced some bias. The MGuard<br />

stent e which has a higher profile and is less flexible than<br />

standard stents e was unable to reach or cross the lesion in<br />

4.1% of patients while there were no device failures in the<br />

control group.<br />

There were no significant differences at 30 days in rates of<br />

mortality (0% versus 1.9%, p ¼ 0.06), major adverse cardiac<br />

events (1.8% versus 2.3%, p ¼ 0.75), or any other clinical outcome.<br />

The authors noted, however, that the study was<br />

underpowered to evaluate differences in clinical events or<br />

infarct size and/or improved clinical outcomes and concluded<br />

that there is need for further experience with this device with<br />

larger trial.<br />

references<br />

1. Abbate A, Kontos MC, Biondi-Zoccai GGL. No-reflow: the next<br />

challenge in treatment of ST-elevation acute myocardial<br />

infarction. Eur <strong>Heart</strong> J. 2008;29(15):1795e1797.<br />

2. Ndrepepa G, Tiroch K, Fusaro M, et al. 5-Year Prognostic Value<br />

of no-reflow phenomenon after percutaneous coronary<br />

intervention in patients with acute myocardial infarction.<br />

J Am Coll Cardiol. 2010;55(21):2383e2389.<br />

3. Stone GW, Abizaid A, Silber S, et al. Prospective, randomized,<br />

multicenter Evaluation of a polyethylene terephthalate<br />

micronet mesh-covered stent (MGuard) in ST-segment<br />

elevation myocardial infarction: the MASTER trial. J Am Coll<br />

Cardiol. 2012;60(19):1975e84.<br />

4. http://www.inspire-md.com/site_en e Official website of<br />

InspireMD Inc.<br />

5. Grube E. New alternative in embolic protection: the mesh<br />

covered MGuard stent. TOUCH BRIEFINGS. Interv Cardiol. 2007:<br />

46e48.<br />

6. Maia F, Ribamar Costa J, Abizaid A, et al. Preliminary results<br />

of the INSPIRE trial with the novel MGuardä stent system<br />

containing a protection net to prevent distal embolization.<br />

Catheter Cardiovasc Interv. 2010;76(1):86e92.<br />

7. Dudek D, Dziewierz A, Rzeszutko q Legutko J, et al. Mesh<br />

covered stent in ST-segment elevation myocardial infarction.<br />

Eurointervention. 2010;6(5):582e589.<br />

8. Danzi A, et al, SOLACI EuroPCR. The International Mguard iMOS<br />

Registry; 2010.<br />

9. Piscione F, Danzi GB, Cassese S, et al. Multicentre experience<br />

with MGuardä net protective stent in ST-elevation<br />

myocardial infarction: safety, feasibility, and impact on<br />

myocardial reperfusion. Catheter Cardiovasc Interv. 2010;75(5):<br />

715e721.<br />

10. The Promise for Life. InspireMD Publication; 2012.<br />

Compiled by<br />

Pankaj V. Jariwala*<br />

Consultant Interventional Cardiologist, Apollo Hospitals,<br />

Hyderguda, Hyderabad, Andhra Pradesh, India<br />

*Tel.: þ91 4064503561, þ91 9393178738 (mobile).<br />

E-mail address: pankaj_jariwala@hotmail.com<br />

M.T. Roe, P.W. Armstrong, K.A. Fox, et al., TRILOGY ACS<br />

Investigators, Prasugrel versus clopidogrel for acute coronary<br />

syndromes without revascularization. N Engl J Med 367 (2012)<br />

1297e1309<br />

1. Background<br />

The effect of intensified platelet inhibition for patients with<br />

unstable angina or myocardial infarction without ST-segment<br />

elevation who do not undergo revascularization has not been<br />

delineated.<br />

2. Methods<br />

In this double-blind, randomized trial, in a primary analysis<br />

involving 7243 patients under the age of 75 years receiving<br />

aspirin, we evaluated upto 30 months of treatment with prasugrel<br />

(10 mg daily) versus clopidogrel (75 mg daily). In a secondary<br />

analysis involving 2083 patients 75 years of age or older,<br />

we evaluated 5 mg of prasugrel versus 75 mg of clopidogrel.<br />

3. Results<br />

At a median follow-up of 17 months, the primary end point of<br />

death from cardiovascular causes, myocardial infarction, or<br />

stroke among patients under the age of 75 years occurred in<br />

13.9% of the prasugrel group and 16.0% of the clopidogrel<br />

group (hazard ratio in the prasugrel group, 0.91; 95% confidence<br />

interval [CI], 0.79 to 1.05; p ¼ 0.21). Similar results were<br />

observed in the overall population. The prespecified analysis


226<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

1000 patients. Primary endpoint at 6 months of MACE of<br />

Ischemia related death, myocardial infarction (Q wave and<br />

non-Q wave), target lesion revascularization (PTCA or CABG).<br />

They enrolled 81% of AMI patients with 77% of STEMI.<br />

Thrombus was visible in 87% of the cases and 65% of the<br />

patients showed initial TIMI flow of 0/1. Device success was<br />

98% and 96% of the patients gained TIMI 3 flow postprocedure.<br />

STEMI subgroup clinical analysis revealed a 30<br />

days accumulative MACE of 2.5% with 0% TLR. 8<br />

As shown in Fig. 4 which compares outcomes of MGuard<br />

stent versus bare metal stent and thrombus aspiration trials<br />

which clearly shows benefit of use of MGuard stent in terms of<br />

blush grade and ST-segment resolution. Though this comparison<br />

is not head to head comparison but trials using bare<br />

metal stent and thrombus aspiration were compared with<br />

similar parameters of myocardial blush grade and ST-segment<br />

resolution. This comparison showed 84.6% MBG grade 3<br />

with MGuard as compared to only 44% with use of thrombus<br />

aspiration catheter and >70% ST resolution was seen in 79.6%<br />

compared to 56.6% in patients of trials of thrombus aspiration.<br />

But these data need careful assessment with statistical analysis<br />

and there is further need of RCTs comparing them. 10<br />

Based on these earlier trials Gregg Stone et al conducted<br />

MASTER trial to prove use of novel stenteMGuard and recommended<br />

in any PCI when a stent implantation is needed<br />

and there is a risk of complication due to distal embolization<br />

of plaque or thrombus. 3<br />

Results of the study showed >70% resolution of STsegment<br />

in MGuard 29% relative to control arm of routine PCI<br />

using either BMS or DES in patients undergoing primary PCI<br />

with thrombus containing lesions but partial or absent resolution<br />

of ST-segment was comparable to control arm.<br />

But this study was limited by the fact that the operators<br />

and research coordinators were not blinded to stent assignment,<br />

which possibly introduced some bias. The MGuard<br />

stent e which has a higher profile and is less flexible than<br />

standard stents e was unable to reach or cross the lesion in<br />

4.1% of patients while there were no device failures in the<br />

control group.<br />

There were no significant differences at 30 days in rates of<br />

mortality (0% versus 1.9%, p ¼ 0.06), major adverse cardiac<br />

events (1.8% versus 2.3%, p ¼ 0.75), or any other clinical outcome.<br />

The authors noted, however, that the study was<br />

underpowered to evaluate differences in clinical events or<br />

infarct size and/or improved clinical outcomes and concluded<br />

that there is need for further experience with this device with<br />

larger trial.<br />

references<br />

1. Abbate A, Kontos MC, Biondi-Zoccai GGL. No-reflow: the next<br />

challenge in treatment of ST-elevation acute myocardial<br />

infarction. Eur <strong>Heart</strong> J. 2008;29(15):1795e1797.<br />

2. Ndrepepa G, Tiroch K, Fusaro M, et al. 5-Year Prognostic Value<br />

of no-reflow phenomenon after percutaneous coronary<br />

intervention in patients with acute myocardial infarction.<br />

J Am Coll Cardiol. 2010;55(21):2383e2389.<br />

3. Stone GW, Abizaid A, Silber S, et al. Prospective, randomized,<br />

multicenter Evaluation of a polyethylene terephthalate<br />

micronet mesh-covered stent (MGuard) in ST-segment<br />

elevation myocardial infarction: the MASTER trial. J Am Coll<br />

Cardiol. 2012;60(19):1975e84.<br />

4. http://www.inspire-md.com/site_en e Official website of<br />

InspireMD Inc.<br />

5. Grube E. New alternative in embolic protection: the mesh<br />

covered MGuard stent. TOUCH BRIEFINGS. Interv Cardiol. 2007:<br />

46e48.<br />

6. Maia F, Ribamar Costa J, Abizaid A, et al. Preliminary results<br />

of the INSPIRE trial with the novel MGuardä stent system<br />

containing a protection net to prevent distal embolization.<br />

Catheter Cardiovasc Interv. 2010;76(1):86e92.<br />

7. Dudek D, Dziewierz A, Rzeszutko q Legutko J, et al. Mesh<br />

covered stent in ST-segment elevation myocardial infarction.<br />

Eurointervention. 2010;6(5):582e589.<br />

8. Danzi A, et al, SOLACI EuroPCR. The International Mguard iMOS<br />

Registry; 2010.<br />

9. Piscione F, Danzi GB, Cassese S, et al. Multicentre experience<br />

with MGuardä net protective stent in ST-elevation<br />

myocardial infarction: safety, feasibility, and impact on<br />

myocardial reperfusion. Catheter Cardiovasc Interv. 2010;75(5):<br />

715e721.<br />

10. The Promise for Life. InspireMD Publication; 2012.<br />

Compiled by<br />

Pankaj V. Jariwala*<br />

Consultant Interventional Cardiologist, Apollo Hospitals,<br />

Hyderguda, Hyderabad, Andhra Pradesh, India<br />

*Tel.: þ91 4064503561, þ91 9393178738 (mobile).<br />

E-mail address: pankaj_jariwala@hotmail.com<br />

M.T. Roe, P.W. Armstrong, K.A. Fox, et al., TRILOGY ACS<br />

Investigators, Prasugrel versus clopidogrel for acute coronary<br />

syndromes without revascularization. N Engl J Med 367 (2012)<br />

1297e1309<br />

1. Background<br />

The effect of intensified platelet inhibition for patients with<br />

unstable angina or myocardial infarction without ST-segment<br />

elevation who do not undergo revascularization has not been<br />

delineated.<br />

2. Methods<br />

In this double-blind, randomized trial, in a primary analysis<br />

involving 7243 patients under the age of 75 years receiving<br />

aspirin, we evaluated upto 30 months of treatment with prasugrel<br />

(10 mg daily) versus clopidogrel (75 mg daily). In a secondary<br />

analysis involving 2083 patients 75 years of age or older,<br />

we evaluated 5 mg of prasugrel versus 75 mg of clopidogrel.<br />

3. Results<br />

At a median follow-up of 17 months, the primary end point of<br />

death from cardiovascular causes, myocardial infarction, or<br />

stroke among patients under the age of 75 years occurred in<br />

13.9% of the prasugrel group and 16.0% of the clopidogrel<br />

group (hazard ratio in the prasugrel group, 0.91; 95% confidence<br />

interval [CI], 0.79 to 1.05; p ¼ 0.21). Similar results were<br />

observed in the overall population. The prespecified analysis


indian heart journal 65 (<strong>2013</strong>) 219e228 227<br />

of multiple recurrent ischemic events (all components of the<br />

primary end point) suggested a lower risk for prasugrel among<br />

patients under the age of 75 years (hazard ratio, 0.85; 95% CI,<br />

0.72e1.00; p ¼ 0.04). Rates of severe and intracranial bleeding<br />

were similar in the two groups in all age groups. There was no<br />

significant between-group difference in the frequency of nonhemorrhagic<br />

serious adverse events, except for a higher frequency<br />

of heart failure in the clopidogrel group.<br />

4. Conclusions<br />

Among patients with unstable nonangina or myocardial<br />

infarction without ST-segment elevation, prasugrel did not<br />

significantly reduce the frequency of the primary end point, as<br />

compared with clopidogrel, and similar risks of bleeding were<br />

observed.<br />

4.1. Clinical perspective<br />

Prasugrel is the more potent alternative antiplatelet drug as<br />

compared to clopidogrel in the treatment of invasively managed<br />

NSTEMI or STEMI as shown in TRITON TIMI 38 in patients<br />

who were treated with invasive strategy. The TRIOLOGY ACS<br />

has tried to answer the use of prasugrel in unstable angina or<br />

NSTEMI patients who are managed medically. The study<br />

population was at high risk, including NSTEMI or UA with<br />

>1 mm of ST depression plus 1 of 4 additional risk criteria:<br />

age 60 years, diabetes, prior MI, prior revascularisation<br />

(percutaneous coronary interventions or coronary artery<br />

bypass grafting). The trial is negative in terms primary end<br />

point of cardiovascular death, MI, or stroke as compared to<br />

clopidogrel. The important limitation of trial was inclusion<br />

criteria as it was not mandatory to do troponin T or I level for<br />

diagnosis and risk stratification of ACS, the troponin T or I<br />

assay was not done in central laboratory, patients were<br />

randomized upto 7 days of acute event and coronary angiography<br />

was not mandatory. The really high risk ACS patient<br />

with proved CAD might have been treated with invasive<br />

strategy and may not have been included in the trial.<br />

There are two important observations from this trial which<br />

has different implications particularly in <strong>Indian</strong> scenario.<br />

The use of Prasugrel in patients who underwent coronary<br />

angiography and had more than 30% coronary stenosis i.e. a<br />

group of patients with definitive CAD but are being managed<br />

conservatively. In this subgroup of patient, there was 23%<br />

reduction in primary end point of cardiovascular death, MI, or<br />

stroke. Thus the patients who have established coronary<br />

artery disease and who are at risk of future cardiac event are<br />

benefited with more potent antiplatelet drug. 1<br />

Second important observation of the trial was potent<br />

antiplatelet activity of prasugrel as compared to clopidogrel.<br />

The platelet function sub study 2 of TRIOLOGY ACS have<br />

shown that the platelet function inhibition was more with<br />

prasugrel than clopidogrel at 30 days among participants<br />

younger than 75 years and weighing 60 kg or more, a significant<br />

difference that persisted through all time points. Thus<br />

it points out that there is no significant difference in ischemic<br />

outcomes through the first 12 months despite clinical observations<br />

of greater P2Y12 inhibition with prasugrel than with<br />

clopidogrel but there is increased risk of bleeding. Thus the<br />

risk of bleeding outweighs the benefit of more potent antiplatelet<br />

activity of prasugrel in treatment of patient subset<br />

included in TRIOLOGY ACS.<br />

In the trial the dose of prasugrel was reduced to 5 mg in<br />

patients more than 75 years of age and body weight less than<br />

60 kg. The question of efficacy of low dose prasugrel have been<br />

tested in recently published FEATHER trial 3 that has shown<br />

that prasugrel 5 mg in low body weight (60 kg), supporting the use of prasugrel 5 mg in<br />

low body weight patients by doing platelet function assay.<br />

In India we have large number of patients who had ACS<br />

who are troponin positive and angiographically proved CAD<br />

but are managed medically because of financial or other reasons.<br />

This subset resembles the angiographically proved CAD<br />

subgroup of TRIOLOGY ACS. Hence the use of body weight<br />

adjusted dose of prasugrel may be extrapolated for conservatively<br />

managed ACS patients who have significant coronary<br />

artery disease and had suffered moderate to high risk<br />

ACS. It will be very interesting to study the medically managed<br />

NSTEMI or UA patients and who have angiographically proven<br />

significant coronary artery disease with use of either prasugrel<br />

or clopidogrel in addition to aspirin. For this the following<br />

study design can be suggested.<br />

Aspirin (75 mg) OD plus Clopidogrel (75 mg)<br />

OD plus standard medical therapy<br />

references<br />

UA or NSTEMI with angiographically<br />

proven CAD & advised PCI/CABGS, but<br />

who have opted for medical management<br />

Aspirin (75mg) OD plus Prasugrel (5 mg)<br />

OD plus standard medical therapy<br />

All patients are followed up for end points like death, non fatal myocardial infarction, heart<br />

failure, stroke and major or minor bleeding or any revascularisation.<br />

1. Wiviott SD. TRILOGY ACS Angiographic Cohort: a Prospective,<br />

Randomized Trial of Prasugrel vs. Clopidogrel in Patients with<br />

Non-ST-Segment-Elevation ACS who are Medically Managed<br />

after Coronary Angiography. TCT 2012; October 24, 2012.<br />

Miami, FL.<br />

2. Gurbel PA, Erlinge D, Ohman EM, et al, For the TRILOGY ACS<br />

platelet function Substudy Investigators. Platelet function<br />

during extended prasugrel and clopidogrel therapy for patients<br />

with ACS treated without revascularization: the TRILOGY ACS<br />

Platelet Function Substudy. JAMA. 2012 Nov 4:1e10.<br />

3. Erlinge D, Berg J, Foley D, et al. Prasugrel 5 mg in low body<br />

weight patient reduces platelet reactivity to a similar extent as<br />

prasugrel 10 mg in higher body weight patients: results from<br />

FEATHER trial. JACC. 2012;60(20):2032e2040.<br />

Parag Admane, N.V. Deshpande<br />

Spandan <strong>Heart</strong> Institute & Research Center, 31,<br />

Off Chitale <strong>Mar</strong>g, Dhantoli, Nagpur 440010, India


228<br />

indian heart journal 65 (<strong>2013</strong>) 219e228<br />

H.M. <strong>Mar</strong>dikar*<br />

Director, Spandan <strong>Heart</strong> Institute & Research Center,<br />

31, Off Chitale <strong>Mar</strong>g, Dhantoli,<br />

Nagpur 440010, India<br />

*Corresponding author. Tel.: þ91 9823082609 (mobile), þ91 (0)<br />

712 2443333; fax: þ91 (0) 712 2443426.<br />

E-mail address: drmardikar@cadindia.co.in<br />

Conclusions: Control patients who crossed over to renal<br />

denervation with the Symplicity system had a significant drop<br />

in blood pressure similar to that observed in patients receiving<br />

immediate denervation. Renal denervation provides safe and<br />

sustained reduction of blood pressure to 1 year.<br />

Clinical trial registration: URL: http://www.clinicaltrials.<br />

gov. Unique identifier: http://www.clinicaltrials.gov. (Circulation.<br />

2012;126:2976e2982.)<br />

Murray D. Esler, Henry Krum, <strong>Mar</strong>kus Schlaich, Roland E.<br />

Schmieder, Michael Böhm, Paul A. Sobotka, for the Symplicity<br />

HTN-2 Investigators. Renal sympathetic denervation for<br />

treatment of drug-resistant hypertension one-year results<br />

from the Symplicity HTN-2 randomized, controlled trial.<br />

Background: Renal sympathetic nerve activation contributes<br />

to the pathogenesis of hypertension. Symplicity HTN-2, a<br />

multicenter, randomized trial, demonstrated that catheterbased<br />

renal denervation produced significant blood pressure<br />

lowering in treatment-resistant patients at 6 months after the<br />

procedure compared with control, medication-only patients.<br />

Longer-term follow-up, including 6-month crossover results,<br />

is now presented.<br />

Methods and results: Eligible patients were on 3 antihypertensive<br />

drugs and had a baseline systolic blood pressure<br />

160 mm Hg (150 mm Hg for type 2 diabetics). After the<br />

6-month primary end point was met, renal denervation in<br />

control patients was permitted. One-year results on patients<br />

randomized to immediate renal denervation (n ¼ 47) and<br />

6-month postprocedure results for crossover patients are<br />

presented. At 12 months after the procedure, the mean fall in<br />

office systolic blood pressure in the initial renal denervation<br />

group ( 28.1 mm Hg; 95% confidence interval, 35.4 to 20.7;<br />

p < 0.001) was similar to the 6-month fall ( 31.7 mm Hg; 95%<br />

confidence interval, 38.3 to 25.0; p < 0.16 versus 6-month<br />

change). The mean systolic blood pressure of the crossover<br />

group 6 months after the procedure was significantly lowered<br />

(from 190.0 19.6 to 166.3 24.7 mm Hg; change, 23.7 27.5;<br />

p < 0.001). In the crossover group, there was 1 renal artery<br />

dissection during guide catheter insertion, before denervation,<br />

corrected by renal artery stenting, and 1 hypotensive<br />

episode, which resolved with medication adjustment.<br />

1. Clinical perspective<br />

The efficacy and safety of the Renal sympathetic Denervation<br />

(RDN) by utilizing the Symplicity Renal Denervation<br />

System in patients with uncontrolled resistant hypertension<br />

have been documented earlier. It has been shown that<br />

activation of the sympathetic nervous system is involved in<br />

the pathogenesis and maintenance of hypertension. Renal<br />

denervation with the Symplicity catheter is a minimally<br />

invasive procedure based on the premise that interruption<br />

of renal afferent and efferent nerves with resultant decrease<br />

in sympathetic outflow to the kidneys should reduce renin<br />

release and sodium retention, increase renal blood flow, and<br />

lower blood pressure. One-year follow-up data of The Symplicity<br />

HTN-2 trial demonstrates two points: (1) that the<br />

initial significant lowering of blood pressure achieved by the<br />

RDN procedure was maintained at the end of one year and<br />

no procedure-related side effect was revealed by that time;<br />

and (2) the control patients maintained on medical therapy<br />

and whose blood pressure was not adequately controlled<br />

were now permitted to switch over to RDN procedure. These<br />

patients again showed significant drop in blood pressure,<br />

which was maintained at the end of six months. Renal<br />

sympathetic Denervation by radiofrequency ablation thus<br />

may provide a safe and effective adjunctive therapy for<br />

treatment-resistant hypertensive patients.<br />

Contributed by:<br />

Arup Dasbiswas<br />

Professor and HOD, Department of Cardiology,<br />

NRS Medical College, Kolkata, India<br />

E-mail address: arup.dasbiswas@gmail.com


indian heart journal 65 (<strong>2013</strong>) 232e233<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Letter to the Editor<br />

Characterization of lipid profile in coronary heart disease<br />

patients in Sudan<br />

Dear Editor,<br />

Although the burden of cardiovascular disease (CVD) states<br />

is stabilizing in high-income countries, in low-to-middleincome<br />

countries it continues to rise. 1 Lifestyle, environmental<br />

and genetic factors play an important role in coronary<br />

heart disease (CHD) development. 2 Previous studies have<br />

shown that blacks have one of the highest rates of coronary<br />

artery disease in the world. 3 However, Caucasians generally<br />

have higher mean total cholesterol (TC) concentrations than<br />

do populations of Asian or African origin. 4<br />

Hospital based case control study was design to study lipid<br />

profile in coronary heart disease patients in Sudan. Among the<br />

total population studied (104 cases and 105 controls) 53.1%<br />

were male, 45% were from Northern Sudan and 72.7% were<br />

residing in urban areas. About 26.8% of the most infected age<br />

group was less than 40 years, 22% had strong family history of<br />

CHD, 42.6% had hypertension and 41.6% had diabetes mellitus.<br />

Smoking and alcohol consumption are very low among<br />

population represent 18.2% and 5.3%, respectively.<br />

Lipid profiles were analyzed using standard enzymatic<br />

methods on a MINDRAY BS-200 analyzer (MINDRAY, Shenzhen,<br />

China). General linear model and correlation between<br />

serum biochemical profiles were performed using SPSS15.0.<br />

The results showed that Sudanese patients had significantly<br />

lower TC and LDL-C levels and non-significantly lower triglycerides,<br />

HDL-C and VLDL levels compared with controls<br />

(Table 1). Age has a significant (p < 0.05) effect on LDL-C, while<br />

sex, race or ethnic, family history, residence, smoking, alcohol<br />

consumption has no significant (p < 0.05) effect on lipid profiles.<br />

Hypertension has no significant (p < 0.05) effect on lipid<br />

profiles, while diabetes mellitus has a significant (p < 0.05)<br />

effect on total cholesterol, and LDL-C (data not shown). Among<br />

patients TC was significantly (p < 0.05) and positively correlated<br />

with LDL and HDL, while VLDL was positively correlated with<br />

triglycerides. In contrast, triglycerides and VLDL were negatively<br />

correlated with LDL and HDL similarly VLDL was positively<br />

correlated with triglycerides (Table 2). In control group<br />

total cholesterol was significantly (p < 0.05) and positively<br />

correlated with triglycerides, LDL and VLDL. Triglyceride was<br />

negatively correlated with HDL and positively with VLDL and<br />

HDL was negatively correlated with LDL and VLDL (Table 2).<br />

Blacks had nominally higher adjusted HDL-C levels, and significantly<br />

lower triglyceride levels than whites. 5 African<br />

ancestry was significantly associated with decreased total<br />

cholesterol, LDL-C and triglycerides. 6 These observed<br />

Table 1 e Baseline characteristics and risk factors for<br />

coronary heart disease in cases and controls subjects.<br />

Traits<br />

Over all<br />

(n ¼ 209)<br />

Case<br />

(n ¼ 104)<br />

Control<br />

(n ¼ 105)<br />

p Value<br />

Age (yrs)<br />

70 (%) 11.5 17.3 5.7 0.014<br />

Ethnic<br />

Northern<br />

45.0 58.7 31.4 0.004<br />

Sudan (%)<br />

Western<br />

34.0 19.2 48.6 0.000<br />

Sudan (%)<br />

Eastern<br />

2.4 2.9 1.9 0.655<br />

Sudan (%)<br />

Southern<br />

1.4 3.8 2.9 0.705<br />

Sudan (%)<br />

Central<br />

17.2 15.4 15.2 1<br />

Sudan (%)<br />

Gender<br />

Male (%) 53.1 56.7 49.5 0.506<br />

Female (%) 46.9 43.3 50.5 0.419<br />

Residence<br />

Urban (%) 72.7 73.1 72.4 1<br />

Rural (%) 27.3 26.9 27.6 0.895<br />

Hypertension (%) 42.6 56.7 28.6 0.002<br />

Diabetes (%) 41.6 40.4 42.9 0.748<br />

Family history (%) 22.0 32.7 11.4 0.001<br />

Smoking (%) 18.2 22.1 14.3 0.196<br />

Alcohol (%) 5.3 7.7 2.9 0.132<br />

TC mg/dL<br />

158.82 9.75 220.90 8.94 0.000<br />

(X SE)<br />

TG mg/dL<br />

243.52 17.23 250.75 15.80 0.749<br />

(X SE)<br />

LDL mg/dL<br />

63.29 8.56 132.22 7.85 0.000<br />

(X SE)<br />

HDL mg/dL<br />

45.91 3.08 46.23 2.82 0.851<br />

(X SE)<br />

VLDL mg/dL<br />

(X SE)<br />

48.78 3.39 51.19 3.11 0.603<br />

TC: total cholesterol mg/dL, TG: triglyceride mg/dL, LDL: low density<br />

lipoprotein mg/dL, HDL: high density lipoprotein mg/dL, VLDL:<br />

very low density lipoprotein mg/dL, CI: 95% confidence interval.<br />

No. of samples for lipid profile for cases and controls were 65 and<br />

78, respectively.


indian heart journal 65 (<strong>2013</strong>) 232e233 233<br />

Table 2 e Correlation coefficient matrixes of cholesterol, triglycerides, and lipoprotein indices in case and control<br />

population.<br />

Traits TC mg/dL TG mg/dL LDL mg/dL HDL mg/dL VLDL mg/dL<br />

TC mg/dL 1 0.214 0.719 a 0.403 a 0.218<br />

TG mg/dL 0.516 a 1 0.070 0.153 0.999 a<br />

LDL mg/dL 0.747 a 0.168 1 0.142 0.065<br />

HDL mg/dL 0.189 0.100 0.039 1 0.153<br />

VLDL mg/dL 0.520 a 0.987 a 0.174 0.103 1<br />

Cases were above the diagonal, and controls were below the diagonal.<br />

a Correlation is significant at the 0.01 level (2-tailed).<br />

associations between African ancestry and several lipid traits<br />

are consistent with the general tendency of individuals of<br />

African descent to have healthier lipid profiles compared to<br />

EuropeaneAmericans. 6 Our results confirm a high prevalence<br />

of conventional risk factors of coronary heart disease as well as<br />

the association between these factors with lipid profiles in<br />

Sudanese population. However, sample size used was very<br />

small of the general population, other studies using large<br />

sample size were needed to provide more accuracy and predictive<br />

value of coronary heart disease risk factors.<br />

Acknowledgments<br />

This study was supported by a grant from the Ministry of<br />

Higher Education, Sudan.<br />

references<br />

1. Abegunde DO, Mathers CD, Adam T, Ortegon M, Strong K. The<br />

burden and costs of chronic diseases in low-income and<br />

middle-income countries. Lancet. 2007;370:1929e1938.<br />

2. Yusuf S, Hawken S, Ounpuu S, et al. Effect of potentially<br />

modifiable risk factors associated with myocardial infarction<br />

in 52 countries (the INTERHEART study): case-control study.<br />

Lancet. 2004;364(9438):937e952.<br />

3. Kountz DS, Levine SL. Cardiovascular risk profiling in blacks:<br />

don’t forget the lipids. Am Fam Physician. 1998;58:1541e1542.<br />

4. Tolonen H, Keil U, Ferrario M, Evans A. Prevalence, awareness<br />

and treatment of hypercholesterolaemia in 32 populations:<br />

results from the WHO MONICA project. Int J Epidemiol. 2005;<br />

34(1):181e192.<br />

5. Aiyer AN, Kip KE, <strong>Mar</strong>roquin OC, Mulukutla SR,<br />

Edmundowicz D, Reis SE. Racial differences in coronary<br />

artery calcification are not attributed to differences in<br />

lipoprotein particle sizes: the <strong>Heart</strong> Strategies Concentrating<br />

on Risk Evaluation (<strong>Heart</strong> SCORE) Study. Am <strong>Heart</strong> J. 2007;153:<br />

328e334.<br />

6. Reiner AP, Carlson CS, Ziv E, Iribarren C, Jaquish CE,<br />

Nickerson DA. Genetic ancestry, population sub-structure,<br />

and cardiovascular disease-related traits among African-<br />

American participants in the CARDIA Study. Hum Genet. 2007;<br />

121:565e575.<br />

Hassan Hussein Musa*<br />

Faculty of Medical Laboratory Sciences, University of Khartoum,<br />

Sudan<br />

Etayeb Mohamed Ahmed Tyrab<br />

Faculty of Medical Laboratory Science, National Ribat University,<br />

Sudan<br />

Muzamil Mahdi Abdel Hamid<br />

Institute of Endemic Diseases, University of Khartoum, Sudan<br />

Elbagire Abdel Rahman Elbashir<br />

Sudan <strong>Heart</strong> Center, Khartoum, Sudan<br />

Lemya Mohammed Yahia, Nihad Mohammed Salih<br />

Faculty of Medical Laboratory Science, National Ribat University,<br />

Sudan<br />

*Corresponding author.<br />

E-mail address: hassan_hm30@yahoo.com<br />

Available online 7 <strong>Mar</strong>ch <strong>2013</strong><br />

0019-4832/$ e see front matter<br />

Copyright ª <strong>2013</strong>, Cardiological Society of India. All rights<br />

reserved.<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.007


indian heart journal 65 (<strong>2013</strong>) 236<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Cardiology News<br />

Dr. Venkat S. Ram awarded Padmashri<br />

Dr. Venkat S. Ram has been awarded<br />

Padmashri recently. He is the<br />

President and CEO of MediCiti Hospitals<br />

and MediCiti Institute of Medical<br />

Sciences, Hyderabad.<br />

He is also Chairman, Board of Governors,<br />

American Society of Hypertension<br />

and Vice-President of<br />

American Society of Hypertension<br />

specialists.<br />

He is a graduate of Osmania Medical College, Hyderabad.<br />

Subsequently, he completed his residency in medicine at<br />

Brown University in Providence, Rhode Island and a fellowship<br />

at the Hospital of the University of Pennsylvania.<br />

Dr. Ram’s professional career has centered on the management<br />

of hypertension and he is one of the very few individuals<br />

who have combined clinical practice with an<br />

academic career.<br />

Dr. Ram served on the Faculty of University of Texas for<br />

many years and was the head of the hypertension clinical<br />

services. He is a prolific writer. He authored more than 310<br />

articles and a book on the treatment of hypertension. He is<br />

also a renowned speaker in the field. Dr. Ram has made<br />

numerous contributions to our understanding of both the<br />

physiology and the management of hypertension. For more<br />

than three decades, Dr. Ram’s work has explored the mechanisms<br />

of action of various antihypertensive drugs. In addition<br />

to expertise in clinical research and medical practice,<br />

Dr. Ram is considered a uniquely skilled communicator of<br />

scientific advances. Dr. Ram was the youngest person ever to<br />

become the Master of American College of Physicians. Dr. Ram<br />

is on the editorial boards of numerous national and international<br />

medical journals. Dr. Ram is now completing a<br />

handbook of hypertension for European use and another book<br />

on hypertension in special populations for Jaypee Brothers<br />

publishers.<br />

Dr. Ram served as the president of American Association of<br />

Physicians from India and also as the president of medical<br />

staff, University Hospital, Dallas, USA. Dr. Ram has been<br />

repeatedly named year after year by the medical students and<br />

residents at the University of Texas Medical School as the<br />

“most skilled teacher of clinical medicine.” The Dallas County<br />

Medical Society, USA has named Dr. Ram as one of the five<br />

individuals in the history of Dallas to make the city as a center<br />

of excellence in medicine. World Telugu Conference bestowed<br />

him the title of the “most outstanding doctor globally of<br />

Telugu origin”daward given by the then Chief Minister, late<br />

N. T. Rama Rao.<br />

He is a founding patron of MediCiti. He conducted several<br />

CME programs and particularly HDL e Hypertension, Diabetes<br />

and Lipids e conferences have been outstanding. Above all he<br />

is a great humanist. Whatever he does as a doctor, specialist,<br />

executive or a friend, it is embellished by his outstanding<br />

humanistic skills. The Cardiological Society of India congratulates<br />

Dr. Venkat S. Ram on receiving this coveted award.<br />

Compiled by<br />

Dr. P.S. Reddy<br />

Hyderabad<br />

0019-4832/$ e see front matter<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.008


indian heart journal 65 (<strong>2013</strong>) 237<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Obituary<br />

The sudden and tragic demise of<br />

Dr Prasanna Nyayadhish on 3rd<br />

January’<strong>2013</strong> has been a devastating<br />

blow for the Cardiology community all<br />

over the country. A brilliant career has<br />

been cut short at its peak, plunging all<br />

his friends, colleagues, and students<br />

into a deep sense of gloom.<br />

Dr Nyayadhish spent his formative<br />

years at the village of Sakharwadi in<br />

district Satara, near Pune. After a bachelor’s degree from BJ<br />

Medical College, Pune in 1992, he received his MD degree in<br />

Medicine from the University of Pune in 1996. Always a brilliant<br />

student, he was one of the top rankers at the All India Superspeciality<br />

Entrance Exam held in 1996. He completed his residency<br />

in DM Cardiology in 2000. He also successfully obtained<br />

the Diplomate of the National Board of Examinations (DNB in<br />

Cardiology). Dr Nyayadhish joined as a full time faculty<br />

member in the Department of Cardiology, KEM Hospital in<br />

2001, eventually rising to the post of Professor and Unit Head in<br />

2008 e a post he held till his demise.<br />

Dr Nyayadhish was an excellent all round Cardiologist. He was<br />

a fine clinician with great powers of observation and analytical<br />

skills. He had a deep knowledge of echocardiography, and was<br />

especially renowned for his abilities in performing and interpreting<br />

echocardiograms in complex congenital heart diseases. In<br />

addition, he was a highly skilled interventionist e performing the<br />

full gamut of cardiac procedures, ranging from balloon atrial<br />

septostomies in one-day-old neonates to complex coronary interventions<br />

in octogenarians. Along with other faculty, he was<br />

responsible for taking Pediatric and Structural heart services at<br />

the KEM hospital to newer heights, making the department one of<br />

its kind in the public health centers in Western India.<br />

Dr Nyayadhish had a friendly and jovial nature. His keen<br />

sense of humor and brilliant repartees made him the life and<br />

soul of gatherings, and his circle of friends extended far<br />

beyond the narrow confines of the medical profession. He was<br />

generous to a fault, warm hearted, and ever ready to help his<br />

colleagues and patients in their times of need. In private<br />

practice, he was respected as much for his ethical and rational<br />

decision making as for his interventional skills.<br />

Dr Nyayadhish was always supportive of research, and<br />

encouraged his residents and faculty to find time from their<br />

clinical work to present and publish research papers. He was a<br />

superb teacher. Along with other senior teachers, he had set up a<br />

teaching academy in the city of Mumbai, for training DM and<br />

DNB students in clinical cardiology, echocardiography, and cardiac<br />

catheterization e all free of charge. The sessions held by this<br />

academy proved to be hugely popular over the last seven to eight<br />

years with trainees all over the country. Dr Nyayadhish was a fine<br />

speaker e his deep knowledge of the subject and unique presentationstylemadehimaregularfixtureasaspeakerorfaculty<br />

in all the important Cardiology conferences and workshops.<br />

The passing of Dr Nyayadhish has been a personal blow for<br />

me e I have lost one of my most valuable faculty members and<br />

a close personal friend. The thoughts of all of us are with his<br />

wife Mrs Lovlyn Nyayadhish and their two young sons in this<br />

difficult time. Let us pray to the Almighty to grant us strength in<br />

bearing this loss. May the departed soul rest in eternal peace.<br />

Compiled by<br />

Dr. Praful Kerkar<br />

Mumbai<br />

0019-4832/$ e see front matter<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.009<br />

Obituary<br />

Dr. N.N. Asokan, a long-standing member of Cardiological Society of India from Muvattupuzha, Kerala expired recently following<br />

a brief illness.<br />

Compiled by<br />

Dr. K. Sarat Chandra<br />

Hony. Editor, <strong>Indian</strong> <strong>Heart</strong> <strong>Journal</strong>, India<br />

0019-4832/$ e see front matter<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.010


indian heart journal 65 (<strong>2013</strong>) 237<br />

Available online at www.sciencedirect.com<br />

journal homepage: www.elsevier.com/locate/ihj<br />

Obituary<br />

The sudden and tragic demise of<br />

Dr Prasanna Nyayadhish on 3rd<br />

January’<strong>2013</strong> has been a devastating<br />

blow for the Cardiology community all<br />

over the country. A brilliant career has<br />

been cut short at its peak, plunging all<br />

his friends, colleagues, and students<br />

into a deep sense of gloom.<br />

Dr Nyayadhish spent his formative<br />

years at the village of Sakharwadi in<br />

district Satara, near Pune. After a bachelor’s degree from BJ<br />

Medical College, Pune in 1992, he received his MD degree in<br />

Medicine from the University of Pune in 1996. Always a brilliant<br />

student, he was one of the top rankers at the All India Superspeciality<br />

Entrance Exam held in 1996. He completed his residency<br />

in DM Cardiology in 2000. He also successfully obtained<br />

the Diplomate of the National Board of Examinations (DNB in<br />

Cardiology). Dr Nyayadhish joined as a full time faculty<br />

member in the Department of Cardiology, KEM Hospital in<br />

2001, eventually rising to the post of Professor and Unit Head in<br />

2008 e a post he held till his demise.<br />

Dr Nyayadhish was an excellent all round Cardiologist. He was<br />

a fine clinician with great powers of observation and analytical<br />

skills. He had a deep knowledge of echocardiography, and was<br />

especially renowned for his abilities in performing and interpreting<br />

echocardiograms in complex congenital heart diseases. In<br />

addition, he was a highly skilled interventionist e performing the<br />

full gamut of cardiac procedures, ranging from balloon atrial<br />

septostomies in one-day-old neonates to complex coronary interventions<br />

in octogenarians. Along with other faculty, he was<br />

responsible for taking Pediatric and Structural heart services at<br />

the KEM hospital to newer heights, making the department one of<br />

its kind in the public health centers in Western India.<br />

Dr Nyayadhish had a friendly and jovial nature. His keen<br />

sense of humor and brilliant repartees made him the life and<br />

soul of gatherings, and his circle of friends extended far<br />

beyond the narrow confines of the medical profession. He was<br />

generous to a fault, warm hearted, and ever ready to help his<br />

colleagues and patients in their times of need. In private<br />

practice, he was respected as much for his ethical and rational<br />

decision making as for his interventional skills.<br />

Dr Nyayadhish was always supportive of research, and<br />

encouraged his residents and faculty to find time from their<br />

clinical work to present and publish research papers. He was a<br />

superb teacher. Along with other senior teachers, he had set up a<br />

teaching academy in the city of Mumbai, for training DM and<br />

DNB students in clinical cardiology, echocardiography, and cardiac<br />

catheterization e all free of charge. The sessions held by this<br />

academy proved to be hugely popular over the last seven to eight<br />

years with trainees all over the country. Dr Nyayadhish was a fine<br />

speaker e his deep knowledge of the subject and unique presentationstylemadehimaregularfixtureasaspeakerorfaculty<br />

in all the important Cardiology conferences and workshops.<br />

The passing of Dr Nyayadhish has been a personal blow for<br />

me e I have lost one of my most valuable faculty members and<br />

a close personal friend. The thoughts of all of us are with his<br />

wife Mrs Lovlyn Nyayadhish and their two young sons in this<br />

difficult time. Let us pray to the Almighty to grant us strength in<br />

bearing this loss. May the departed soul rest in eternal peace.<br />

Compiled by<br />

Dr. Praful Kerkar<br />

Mumbai<br />

0019-4832/$ e see front matter<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.009<br />

Obituary<br />

Dr. N.N. Asokan, a long-standing member of Cardiological Society of India from Muvattupuzha, Kerala expired recently following<br />

a brief illness.<br />

Compiled by<br />

Dr. K. Sarat Chandra<br />

Hony. Editor, <strong>Indian</strong> <strong>Heart</strong> <strong>Journal</strong>, India<br />

0019-4832/$ e see front matter<br />

http://dx.doi.org/10.1016/j.ihj.<strong>2013</strong>.03.010

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!