15.02.2015 Views

THE JOURNAL OF TEHRAN UNIVERSITY HEART CENTER

THE JOURNAL OF TEHRAN UNIVERSITY HEART CENTER

THE JOURNAL OF TEHRAN UNIVERSITY HEART CENTER

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Zohair Yousef Al-halees, MD , FRCSC, FACS<br />

King Faisal Heart Institute, Saudi Arabia<br />

Yadolah Dodge, PhD<br />

University of Neuchâtel, Switzerland<br />

Ali Dodge–Khatami, MD, PhD<br />

University of Hamburg- Eppendorf School of Medicne, Germany<br />

Iradj Gandjbakhch, MD<br />

Hopital Pitie, France<br />

Omer Isik, MD<br />

Yeditepe University, School of Medicine, Turkey<br />

Sami S. Kabbani, MD<br />

Damascus University Cardiovascular Surgical Center, Syria<br />

Kayvan Kamalvand, MD, FRCP, FACC<br />

William Harvey Hospital, United Kingdom<br />

Jean Marco, MD, FESC<br />

Centre Cardio- Thoracique de Monaco, France<br />

Ali Massumi, MD<br />

Texas Heart Institute, U. S. A<br />

Carlos-A. Mestres, MD<br />

University of Barcelona, Spain<br />

Hossien Ahmadi, MD<br />

Tehran University of Medical Sciences<br />

Shahin Akhondzadeh, PhD<br />

Tehran University of Medical Sciences<br />

Mohammad Alidoosti, MD<br />

Tehran University of Medical Sciences<br />

Mohammad Ali Boroumand, MD<br />

Tehran University of Medical Sciences<br />

Gholamreza Davoodi, MD<br />

Tehran University of Medical Sciences<br />

Ahmad Reza Dehpour, PhD<br />

Tehran University of Medical Sciences<br />

Alimohammad Haji Zeinali, MD<br />

Tehran University of Medical Sciences<br />

Abbasali Karimi, MD<br />

Tehran University of Medical Sciences<br />

Ali Kazemi Saeed, MD<br />

Tehran University of Medical Sciences<br />

Seyed Ebrahim Kassaian, MD<br />

Tehran University of Medical Sciences<br />

Davood Kazemi Saleh, MD<br />

Baghiatallah University of Medical Sciences<br />

Majid Maleki, MD<br />

Tehran University of Medical Sciences<br />

Mehrab Marzban, MD<br />

Shahid Beheshti University of Medical Sciences<br />

<strong>THE</strong><br />

<strong>JOURNAL</strong> <strong>OF</strong> <strong>TEHRAN</strong><br />

<strong>UNIVERSITY</strong> <strong>HEART</strong><br />

<strong>CENTER</strong><br />

Editor-in-Chief<br />

ABBASALI KARIMI, MD<br />

PR<strong>OF</strong>ESSOR <strong>OF</strong> CARDIAC SURGERY<br />

<strong>TEHRAN</strong> <strong>UNIVERSITY</strong> <strong>OF</strong> MEDICAL SCIENCES<br />

Associate & Managing Editor<br />

SEYED HESAMEDDIN ABBASI, MD<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

<strong>TEHRAN</strong> <strong>UNIVERSITY</strong> <strong>OF</strong> MEDICAL SCIENCES<br />

International Editors<br />

Editorial Board<br />

Fred Morady, MD<br />

University of Michigan, U. S. A<br />

Mohammed T. Numan, MD<br />

University of Texas, U. S. A<br />

Ahmand S. Omran, MD, FACC, FASE<br />

King Abdulaziz Cardiac Center, Saudi Arabia<br />

Fausto J. Pinto, MD, PhD, FESC, FACC, FASA, FSCAI,<br />

FASE<br />

Lisbon University, Portugal<br />

Mehrdad Rezaee, MD, PhD<br />

Stanford University, School of Medicine, U. S. A<br />

Gregory S. Thomas, MD, MPH, FACC, FACP, FASNC<br />

University of California, U. S. A<br />

Lee Samuel Wann, MD<br />

Wisconsin Heart Hospital, U. S. A<br />

Hein J. Wellens, MD<br />

Cardiovascular Research Institute, Maastricht, The Netherlands<br />

Douglas P. Zipes, MD<br />

Indiana University, School of medicine, U. S. A<br />

Mansor Moghadam, MD<br />

Tehran University of Medical Sciences<br />

Sina Moradmand Badie, MD<br />

Tehran University of Medical Sciences<br />

Seyed Mahmood Mirhoseini, MD, DSc, FACC, FAES<br />

Tehran University of Medical Sciences<br />

Seyed Rasoul Mirsharifi, MD<br />

Tehran University of Medical Sciences<br />

Ahmad Mohebi, MD<br />

Tehran University of Medical Sciences<br />

Mohammad-Hasan Namazi<br />

Shaheed beheshti University of Medical Sciences<br />

Ebrahim Nematipour, MD<br />

Tehran University of Medical Sciences<br />

Rezayat Parvizi, MD<br />

Tabriz University of Medical Sciences<br />

Masoud Pezeshkian<br />

Tabriz University of Medical Sciences<br />

Hamid Reza Pour Hosseini, MD<br />

Tehran University of Medical Sciences<br />

Hassan Radmehr, MD<br />

Tehran University of Medical Sciences<br />

Hakimeh Sadeghian, MD<br />

Tehran University of Medical Sciences<br />

Saeed Sadeghian, MD<br />

Tehran University of Medical Sciences


Mojtaba Salarifar, MD<br />

Tehran University of Medical Sciences<br />

Nizal Sarraf –Zadegan, MD<br />

Isfahan University of Medical Sciences<br />

Ahmad Yaminisharif, MD<br />

Tehran University of Medical Sciences<br />

Kiyomars Abbasi, MD<br />

Tehran University of Medical Sciences<br />

Seifollah Abdi, MD<br />

Tehran University of Medical Sciences<br />

Hassan Aghajani, MD<br />

Tehran University of Medical Sciences<br />

Alireza Amirzadegan, MD<br />

Tehran University of Medical Sciences<br />

Naser Aslanabadi, MD<br />

Tabriz University of Medical Sciences<br />

Carlo Briguori, MD<br />

Clinica Mediterranea Hospital, Naples, Italy<br />

Alaide Chieffo, MD<br />

San Raffaele Scientific Institute, Milano, Italy<br />

Sirous Darabian, MD<br />

St. John Cardiovascular Reserach Center, USA<br />

Saeed Davoodi, MD<br />

Tehran University of Medical Sciences<br />

Iraj Firoozi, MD<br />

Tehran University of Medical Sciences<br />

Seyed Khalil Foroozannia, MD<br />

Shaheed Sadoghi University of Medical Sciences<br />

Alfredo R. Galassi MD, FACC, FESC, FSCAI<br />

Cannizzaro Hospital, Catania, Italy<br />

Armen Gasparyan MD, PhD<br />

Armenia<br />

Ali Ghaemian, MD<br />

Mazandaran University of Medical Sciences<br />

Namvar Ghasemi Movahedi, MD<br />

Tehran University of Medical Sciences<br />

Abbas Ghiasi, MD<br />

Tehran University of Medical Sciences<br />

Mahmood Sheikh Fathollahi, PhD<br />

Arash Jalali, PhD<br />

Farshad Amouzadeh<br />

Fatemeh Esmaeili Darabi<br />

Advisory Board<br />

Statistical Consultant<br />

Technical Editors<br />

Graphic Design & Office<br />

Mohammad Reza Zafarghandi, MD<br />

Tehran University of Medical Sciences<br />

Aliakbar Zeinaloo, MD<br />

Tehran University of Medical Sciences<br />

Mohammad Jafar Hashemi, MD<br />

Tehran University of Medical Sciences<br />

Seyed Kianoosh Hoseini<br />

Tehran University of Medical Sciences<br />

Elise Langdon- Neuner<br />

European Medical Writers Association, Austria<br />

Jalil Majd Ardekani, MD<br />

Tehran University of Medical Sciences<br />

Fardin Mirbolook, MD<br />

Gilan University of Medical Sciences<br />

Mehdi Najafi, MD<br />

Tehran University of Medical Sciences<br />

Younes Nozari, MD<br />

Tehran University of Medical Sciences<br />

Mohammad Saheb Jam, MD & PT<br />

Tehran University of Medical Sciences<br />

Abbas Salehi Omran, MD<br />

Tehran University of Medical Sciences<br />

Mahmood Shabestari, MD<br />

Mashhad University of Medical Sciences<br />

Shapour Shirani, MD<br />

Tehran University of Medical Sciences<br />

Abbas Soleimani, MD<br />

Tehran University of Medical Sciences<br />

Seyed Abdolhosein Tabatabaei, MD<br />

Tehran University of Medical Sciences<br />

Murat Ugurlucan, MD<br />

Duzce Ataturk State Hospital, Turkey<br />

Arezou Zoroufian, MD<br />

Tehran University of Medical Sciences<br />

The Journal of Tehran University Heart Center is indexed in PubMed, PubMed Central, Scopus, EMBASE,<br />

CAB Abstracts, Global Health, Chemical Abstract Service, Cinahl, ProQuest, Google Scholar, DOAJ, EBSCO,<br />

Geneva Foundation for Medical Education and Research, Index Copernicus, Index Medicus for the WHO Eastern<br />

Mediterranean Region (IMEMR), ISC, SID, Iranmedex and Magiran<br />

Address<br />

North Kargar Street, Tehran Heart Center, Tehran, Iran. 1411713138. Tel: +98-21-88029720. Fax: +98-21-88029702.<br />

Web Site: http://jthc.tums.ac.ir. E-mail: jthc@tums.ac.ir.


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Content<br />

Volume: 7 Number: 4 Autumn 2012<br />

The Journal of Tehran University Heart Center<br />

Review Article<br />

Prosthetic Tricuspid Valve Thrombosis: Three Case Reports and Literature Review<br />

Ahmad Yaminisharif, Mohammad Javad Alemzadeh-Ansari, Seyed Hossein Ahmadi ……………..…........................................................................... 147<br />

Original Articles<br />

Increased Carotid Artery Intima-Media Thickness in Pregnant Women with Gestational Diabetes Mellitus<br />

Gholamreza Yousefzadeh, Hashem Hojat, Ahmad Enhesari, Mostafa Shokoohi, Nahid Eftekhari, Mehrdad Sheikhvatan …………….....………………. 156<br />

Right Ventricular Myocardial Tissue Velocities, Myocardial Performance Index, and Tricuspid Annular Plane<br />

Systolic Excursion in Totally Corrected Tetralogy of Fallot Patients<br />

Asadolah Tanasan, Keyhan Sayadpour Zanjani, Armen Kocharian, Abdolrazagh Kiani, Mohammad Ali Navabi ……………...……............................ 160<br />

Anatomy of Atrioventricular Node Artery and Pattern of Dominancy in Normal Coronary Subjects: A<br />

Comparison Between Individuals With and Without Isolated Right Bundle Branch Block<br />

Ali Kazemisaeid, Marziyeh Pakbaz, Ahmad Yaminisharif, Gholamreza Davoodi, Masoumeh Lotfi Tokaldany, Elham Hakki Kazazi ……..................... 164<br />

Determinants of Length of Stay in Surgical Ward after Coronary Bypass Surgery: Glycosylated Hemoglobin as<br />

a Predictor in All Patients, Diabetic or Non-Diabetic<br />

Mahdi Najafi, Hamidreza Goodarzynejad …………….......………..........................….................................................................................................... 170<br />

Obvious or Subclinical Right Ventricular Dysfunction in Diabetes Mellitus (Type II): An Echocardiographic<br />

Tissue Deformation Study<br />

Mozhgan Parsaee, Parvaneh Bahmanziari, Maryam Ardeshiri, Maryam Esmaeilzadeh ……...........….....................................................................…… 177<br />

Case Reports<br />

Surgical Treatment of Amplatzer Embolus in a Secundum Atrial Septal Defect Patient<br />

Ahmet Baris Durukan, Hasan Alper Gurbuz, Murat Tavlasoglu, Nevriye Salman, Halil Ibrahim Ucar, Cem Yorgancioglu ………….……….............. 182<br />

Post-Traumatic Chordae Rupture of Tricuspid Valve<br />

Kyomars Abbasi, Hossein Ahmadi, Arezoo Zoroufian, Mohammad Sahebjam, Naghmeh Moshtaghi, Seyed Hessamedin Abbasi ................................ 185<br />

Late Diagnosis of Large Left Ventricular Pseudoaneurysm after Mitral Valve Replacement and Coronary Artery<br />

Bypass Surgery by Real-Time Three-Dimensional Echocardiography<br />

Mohammad Sahebjam, Abbas Salehiomran, Neda Ghaffari-Marandi, Azam Safir …………….....………..…............................................................... 188<br />

Letter to the Editor<br />

A Memorandum of "World Heart Day 2012": Myocardial Infarction Mortality in Women in Birjand, 2008-2009<br />

Toba Kazemi, Gholam Reza Sharifzadeh …………........................................................................................................................................................... 191<br />

Photo Clinic<br />

Ball in Chest<br />

Mohammad Bagher Rahim, Mohammad Hossein Mandegar, Farideh Roshanali …………............................................................................................. 192<br />

The Journal of Tehran University Heart Center


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong>


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Volume: 7 Number: 4<br />

Summary of Contents<br />

What you will find<br />

in this Issue<br />

Autumn 2012<br />

REVIEW ARTICLE<br />

147 Prosthetic Tricuspid Valve Thrombosis:<br />

Three Case Reports and Literature Review<br />

Ahmad Yaminisharif, MD, Mohammad Javad Alemzadeh-<br />

Ansari, MD * , Seyed Hossein Ahmadi, MD<br />

Tehran Heart Center, Tehran University of Medical Sciences,<br />

Tehran, Iran.<br />

*<br />

Mohammad Javad Alemzadeh-Ansari, Department of<br />

Cardiology, Tehran University of Medical Sciences, Tehran<br />

Heart Center, Karegar Street, Tehran, Iran. 1411713138.<br />

Tel: +98 21 88029600. Fax: +98 21 88029731. E-mail:<br />

aansari@razi.tums.ac.ir.<br />

common complication of prosthetic heart<br />

A valves is thrombosis. Although the incidence<br />

of prosthetic valve thrombosis (PVT) in the tricuspid<br />

position is high, there are not enough data on the<br />

management of it, in contrast to left-sided PVT.<br />

Here, we describe three cases of tricuspid PVT with<br />

three different management approaches: thrombolytic<br />

therapy; close observation with oral anticoagulants;<br />

and surgery. The first case was a woman who suffered<br />

from recurrent PVT, for which we successfully used<br />

Tenecteplase for second and third episodes. We<br />

employed Tenecteplase in this case for the first time in<br />

the therapy of tricuspid PVT. The second case had fixed<br />

leaflets in open position while being symptomless. At<br />

six months' follow-up, with the patient having taken<br />

oral anticoagulants, the motion of the leaflets was<br />

restricted and she was symptom-free. The last case<br />

was a woman who had a large thrombus in the right<br />

atrium immediately after mitral and tricuspid valvular<br />

replacement. The patient underwent re-replacement<br />

surgery and a new biological valve was implanted in<br />

the tricuspid position. Also, we review the literature<br />

on the pathology, signs and symptoms, diagnosis, and<br />

management of tricuspid PVT.<br />

ORIGINAL ARTICLES<br />

156 Increased Carotid Artery Intima-Media<br />

Thickness in Pregnant Women with<br />

Gestational Diabetes Mellitus<br />

GholamrezaYousefzadeh, MD 1 , Hashem Hojat, MD 1 , Ahmad<br />

Enhesari, MD 1 , Mostafa Shokoohi, MSc 2* , Nahid Eftekhari,<br />

MD 1 , Mehrdad Sheikhvatan, MD 3<br />

1<br />

Physiology Research Center, Kerman University of Medical<br />

Sciences, Kerman, Iran.<br />

2<br />

Research Center for Modeling in Health, Kerman University<br />

of Medical Sciences, Kerman, Iran.<br />

3<br />

Tehran Heart Center, Tehran University of Medical Sciences,<br />

Tehran, Iran.<br />

*<br />

Mostafa Shokoohi, Research Center for Modeling in Health,<br />

Kerman University of Medical Sciences, Jihad Blvd, Shariati Street,<br />

Azadi Square, Kerman, Iran. 7619813159. Tel: +98 341 2263983.<br />

Fax: +98 341 2264079. E-mail: shokouhi.mostafa@gmail.com.<br />

Background: Pregnant women with previous<br />

gestational diabetes mellitus are at increased<br />

risk of progressive carotid artery disorders. The current<br />

study evaluated carotid intima-media thickness (IMT)<br />

in pregnant women with gestational diabetes at two<br />

time points of mid-term and full-term pregnancy to<br />

determine whether gestational diabetes mellitus causes<br />

increased IMT.<br />

Conclusion: In conclusion, an impaired OGCT<br />

test is proven to be an independent risk factor for<br />

increased carotid IMT and subsequent coronary artery<br />

disease. Even with this small study, we were able to<br />

find an increased IMT after diabetes appearance, which<br />

might be used as an indicator of a potential increased<br />

vascular risk. Furthermore, IMT measurements in<br />

diabetic pregnant women could offer an opportunity<br />

to identify a high-risk group of women who might


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

benefit from early screening and preventive measures.<br />

These measures could include lifestyle interventions<br />

such as improving diet and physical activity as well as<br />

increased surveillance of blood pressure, serum lipids,<br />

and particularly blood glucose.<br />

160 Right Ventricular Myocardial Tissue<br />

Velocities, Myocardial Performance<br />

Index, and Tricuspid Annular Plane<br />

Systolic Excursion in Totally Corrected<br />

Tetralogy of Fallot Patients<br />

Asadolah Tanasan, MD 1 , Keyhan Sayadpour Zanjani,<br />

MD 2* , Armen Kocharian, MD 2 , Abdolrazagh Kiani, MD 2 ,<br />

Mohammad Ali Navabi, MD 2<br />

1<br />

Besat Hospital, Hamadan University of Medical Sciences,<br />

Hamadan, Iran.<br />

2<br />

Children’s Medical Center, Tehran University of Medical<br />

Sciences, Tehran, Iran.<br />

*<br />

Keyhan Sayadpour Zanjani, Assistant Professor of Pediatric<br />

Cardiology, Tehran University of Medical Sciences, Children’s<br />

Medical Center, No.62, Dr Gharib Street, Tehran, Iran. 1419733151.<br />

Tel: +98 21 66911029. Fax: +98 21 66930024. E-mail: sayadpour@<br />

tums.ac.ir.<br />

Background: Longer survival after the total<br />

repair of the Tetralogy of Fallot increases<br />

the importance of late complications such as right<br />

ventricular dysfunction. This is a prospective study<br />

of the right ventricular function in totally corrected<br />

Tetralogy of Fallot patients versus healthy children.<br />

Conclusion: RVMPI was significantly correlated<br />

with PR severity without the presence of a significant<br />

correlation between RVMPI (obtained by pulsed wave<br />

Doppler) and the RV function indices obtained by tissue<br />

Doppler imaging (EA, Aa, Ea/Aa, and Sa). We suggest<br />

that these indices, RVMPI by tissue Doppler and PRi,<br />

be measured basically at postoperative and follow-up<br />

evaluations. In addition, as TAPSE was significantly<br />

decreased in the totally corrected T<strong>OF</strong> patients and<br />

there was a significant correlation between TAPSE and<br />

Sa, we suggest that TAPSE be also measured as the<br />

global RV systolic function index.<br />

164 Anatomy of Atrioventricular Node Artery<br />

and Pattern of Dominancy in Normal<br />

Coronary Subjects: A Comparison between<br />

Individuals with and without Isolated<br />

Right Bundle Branch Block<br />

Ali Kazemisaeid, MD, Marziyeh Pakbaz, MD * , Ahmad<br />

Yaminisharif, MD, Gholamreza Davoodi, MD, Masoumeh<br />

Lotfi Tokaldany, MD, Elham Hakki Kazazi, MD<br />

Tehran Heart Center, Tehran University of Medical Sciences,<br />

Tehran, Iran.<br />

*<br />

Marziyeh Pakbaz, Department of Cardiology, Tehran University<br />

of Medical Sciences, Tehran Heart Center, North Kargar Street,<br />

Tehran, Iran.1411713138. Tel: +98 21 88029256. Fax: +98 21<br />

88029256. E-Mail: marzi.pakbaz@gmail.com.<br />

Background: Isolated right bundle branch block<br />

(RBBB) is a common finding in the general<br />

population. The atrioventricular node (AVN) artery<br />

contributes to the blood supply of the right bundle<br />

branch. Our hypothesis was that the anatomy of<br />

the AVN artery and the pattern of dominancy differ<br />

between subjects with and without RBBB.<br />

Conclusion: According to our observations, there<br />

was no relationship between the dominancy of the<br />

epicardial arteries and the presence of RBBB in<br />

subjects with normal coronary arteries. There was<br />

a great variation of the AVN artery origin. Non-crux<br />

origination of the AVN artery was more common than<br />

the crux origination in both groups, and the prevalence<br />

of non-crux origination of the AVN artery was<br />

significantly higher in the cases than in the controls.<br />

Origination of the AVN artery from the right circulatory<br />

system was more common in both groups and the<br />

prevalence of the right origin of the AVN artery was<br />

significantly higher in the cases than in the controls.<br />

The AVN artery most commonly originated from the<br />

dominant artery but not necessarily from the crux.<br />

170 Determinants of Length of Stay in Surgical<br />

Ward after Coronary Bypass Surgery:<br />

Glycosylated Hemoglobin as a Predictor in<br />

All Patients, Diabetic or Non-Diabetic<br />

Mahdi Najafi, MD * , Hamidreza Goodarzynejad, MD


1<br />

Tehran Heart Center, Tehran University of Medical Sciences,<br />

Tehran, Iran.<br />

*<br />

Mahdi Najafi, Assistant Professor of Anesthesiology, Tehran<br />

Heart Center, Tehran University of Medical Sciences, Tehran Heart<br />

Center, North Karegar Street, Tehran, Iran. 1411713138. Tel: +98 21<br />

88029600. Fax: +98 21 88029731. E-mail: najafik@sina.tums.ac.ir.<br />

Background: Reports on the determinants of<br />

morbidity in coronary artery bypass graft surgery<br />

(CABG) have focused on outcome measures such as<br />

length of postoperative stay in the Intensive Care Unit<br />

(ICU). We proposed that major comorbidities in the<br />

ICU hampered the prognostic effect of other weaker<br />

but important preventable risk factors with effect on<br />

patients’ length of hospitalization. So we aimed at<br />

evaluating postoperative length of stay in the ICU and<br />

surgical ward separately.<br />

Conclusion: Among the five predictors of the LOS<br />

in the surgical ward, three were the indices of glycemic<br />

control. HbA1c is the most reliable marker of outcome<br />

because its level is not influenced by perioperative<br />

events. Taking these findings into consideration,<br />

the prognostic role of HbA1c for the LOS following<br />

CABG is promising. We would, therefore, recommend<br />

its use as a simple predictor of outcome after cardiac<br />

surgery in daily practice.<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Background: Diabetes mellitus is capable of<br />

impairing the myocardial function. Several<br />

studies have documented the influential impact of<br />

diabetes mellitus on the left ventricular function. The<br />

right ventricular function plays a significant role in the<br />

overall myocardial contractility; hence, this study was<br />

undertaken to evaluate the effect of diabetes mellitus<br />

type II on the right ventricular function.<br />

Conclusion: Whether the RV diastolic abnormalities<br />

have prognostic implications in the clinical course of<br />

patients with DM type II remains to be investigated. We<br />

believe that serial echocardiography measurements are<br />

warranted in this diabetic population if the progression<br />

from subclinical RV involvement to symptomatic RV<br />

dysfunction is to be followed. In addition, subclinical<br />

RV systolic and diastolic abnormalities should be<br />

considered when planning pharmacotherapy to prevent<br />

the development of symptomatic RV dysfunction.<br />

CASE REPORTS<br />

182 Surgical Treatment of Amplatzer Embolus<br />

in a Secundum Atrial Septal Defect Patient<br />

Ahmet Baris Durukan, MD 1* , Hasan Alper Gurbuz, MD 1 ,<br />

Murat Tavlasoglu, MD 2 , Nevriye Salman, MD 1 , Halil Ibrahim<br />

Ucar, MD 1 , Cem Yorgancioglu, MD 1<br />

1<br />

Medicana International Ankara Hospital, Ankara, Turkey.<br />

2<br />

Diyarbakir Military Hospital, Diyarbakir, Turkey.<br />

177 Obvious or Subclinical Right Ventricular<br />

Dysfunction in Diabetes Mellitus (Type II):<br />

An Echocardiographic Tissue Deformation<br />

Study<br />

Mozhgan Parsaee, MD * , Parvaneh Bahmanziari, MD,<br />

Maryam Ardeshiri, MD, Maryam Esmaeilzadeh, MD<br />

Rajaei Cardiovascular, Medical and Research Center, Tehran<br />

University of Medical Sciences, Tehran, Iran.<br />

*<br />

Mozhgan Parsaee, Assistant Professor of Cardiology,<br />

Department of Echocardiography, Rajaei Cardiovascular, Medical<br />

and Research Center, Vali-Asr Ave. Adjacent to Mellat Park, Tehran,<br />

Iran. 1996911151. Tel: +98 21 23922930. Fax: +98 21 2055594.<br />

E-mail: parsaeemozhgan@yahoo.com.<br />

*<br />

Ahmet Baris Durukan, Medicana International Ankara<br />

Hospital, Umit Mahallesi 2463.sokak 4/18, 06810, Yenimahalle,<br />

Ankara, Turkey. Tel: +90 532 2273814. Fax: +90 312 2203170.<br />

E-mail: barisdurukan@yahoo.com.<br />

Transcatheter device closure of the ASD can lead<br />

to serious complications despite its advantages<br />

over surgical closure. But it should be kept in mind<br />

that even if most cases are successful, not only early,<br />

but also late complications may occur regardless<br />

of the size or type of the current devices. Surgical<br />

treatment of the complications may be mandatory<br />

and should be performed immediately, especially<br />

in cases of embolization. Surgery is quite effective<br />

in treatment, but it is a fact that operative mortality<br />

rises when surgery is performed for the treatment of<br />

complications. Operative mortality is much lower for<br />

primary surgical repair.


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

185 Post-Traumatic Chordae Rupture of<br />

Tricuspid Valve<br />

Kyomars Abbasi, MD, Hossein Ahmadi, MD, Arezoo<br />

Zoroufian, MD, Mohammad Sahebjam, MD, Naghmeh<br />

Moshtaghi, MD, Seyed Hessamedin Abbasi, MD *<br />

Tehran Heart Center, Tehran University of Medical Sciences,<br />

Tehran, Iran.<br />

*<br />

Seyed-Hesameddin Abbasi, Tehran University of<br />

Medical Sciences, Tehran Heart Center, North Kargar Street,<br />

Tehran, Iran. 1411713138. Tel: +98 21 88029720. Fax: +98 21<br />

88029702. E-mail: abbasi@tehranheartcenter.org.<br />

The two cases presented herein demonstrate<br />

that the rupture of the chordae tendineae of the<br />

tricuspid valve could be another cause of TR following<br />

blunt chest trauma. This is, however, a condition that<br />

can be repaired surgically without the need for valve<br />

replacement. Advances in echocardiography have<br />

enabled earlier diagnosis and ergo more effective<br />

treatment.<br />

We recommend that physicians working at<br />

emergency departments be on the alert for this<br />

potential complication of non-penetrating chest trauma<br />

and subject all patients admitted to the emergency<br />

department due to blunt chest trauma to TTE or TEE<br />

for accurate diagnosis.<br />

The LV pseudoaneurysm is an important<br />

complication that may occur late after MVR and<br />

thus necessitates due heed on the part of cardiologists<br />

and cardiac surgeons.<br />

LETTER TO <strong>THE</strong> EDITOR<br />

191 A Memorandum of "World Heart Day<br />

2012": Myocardial Infarction Mortality in<br />

Women in Birjand, 2008-2009<br />

Toba Kazemi, MD, Gholam Reza Sharifzadeh, MSc<br />

Valiassr Hospital, Birjand University of Medical Sciences<br />

(BUMS), Birjand, Iran.<br />

*<br />

Toba Kazemi, Associate Professor of Cardiology, Birjand<br />

University of Medical Sciences (BUMS), Valiassr Hospital, Ghafari<br />

Avenue, Birjand, Iran. 9717964151. Tel: +98 56 14443001-9. Fax:<br />

+98 56 14433004. E-mail: drtooba.kazemi@gmail.com.<br />

192 Ball in Chest<br />

PHOTO CLININC<br />

Mohammad Bagher Rahim, MD, Mohammad Hossein<br />

Mandegar, MD, Farideh Roshanali, MD *<br />

Shariati General Hospital, Tehran University of Medical<br />

Sciences, Tehran, Iran.<br />

188 Late Diagnosis of Large Left Ventricular<br />

Pseudoaneurysm after Mitral Valve<br />

Replacement and Coronary Artery Bypass<br />

Surgery by Real-Time Three-Dimensional<br />

Echocardiography<br />

*<br />

Farideh Roshanali, Shriati Hospital, North Kargar Street,<br />

Tehran, Iran. Tel: +98 912 3093151. Fax: +98 21 88797353. E-mail:<br />

farideh_roshanali@yahoo.com.<br />

Mohammad Sahebjam, MD * , Abbas Salehiomran, MD, Neda<br />

Ghaffari-Marandi, MD, Azam Safir, MD<br />

Tehran Heart Center, Tehran University of Medical Sciences,<br />

Tehran, Iran.<br />

*<br />

Mohammad Sahebjam, Assistant Professor of Cardiology,<br />

Echocardiography Department, Tehran Heart Center, Jalal Al Ahmad<br />

and North Kargar Intersection, Tehran, Iran. 1411713138. Tel: +98 21<br />

88029600. Fax: +98 21 88029731. E-mail: msahebjam@yahoo.com.


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Review Article<br />

Prosthetic Tricuspid Valve Thrombosis: Three Case Reports<br />

and Literature Review<br />

Ahmad Yaminisharif, MD, Mohammad Javad Alemzadeh-Ansari, MD * ,<br />

Seyed Hossein Ahmadi, MD<br />

Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.<br />

Received 24 January 2012; Accepted 17 June 2012<br />

Abstract<br />

A common complication of prosthetic heart valves is thrombosis. Although the incidence of prosthetic valve thrombosis<br />

(PVT) in the tricuspid position is high, there are not enough data on the management of it, in contrast to left-sided PVT.<br />

Here, we describe three cases of tricuspid PVT with three different management approaches: thrombolytic therapy; close<br />

observation with oral anticoagulants; and surgery. The first case was a woman who suffered from recurrent PVT, for which<br />

we successfully used Tenecteplase for second and third episodes. We employed Tenecteplase in this case for the first time in<br />

the therapy of tricuspid PVT. The second case had fixed leaflets in open position while being symptomless. At six months'<br />

follow-up, with the patient having taken oral anticoagulants, the motion of the leaflets was restricted and she was symptomfree.<br />

The last case was a woman who had a large thrombus in the right atrium immediately after mitral and tricuspid valvular<br />

replacement. The patient underwent re-replacement surgery and a new biological valve was implanted in the tricuspid<br />

position. Also, we review the literature on the pathology, signs and symptoms, diagnosis, and management of tricuspid PVT.<br />

J Teh Univ Heart Ctr 2012;7(4):147-155<br />

This paper should be cited as: Yaminisharif A, Alemzadeh-Ansari MJ, Ahmadi SH. Prosthetic Tricuspid Valve Thrombosis: Three<br />

Case Reports and Literature Review. J Teh Univ Heart Ctr 2012;7(4):147-155.<br />

Keywords: Tricuspid valve • Thrombosis • Thrombolytic therapy • Anticoagulants • Surgical procedures, operative<br />

Introduction<br />

Since the 1950s, more than 80 models of the prosthetic<br />

heart valve have been developed and used. 1 Prosthetic valve<br />

thrombosis (PVT), however, remains a serious complication<br />

and can even prove lethal. Overall, the incidence of<br />

thrombosis is reported to be between 0.1% and 5.7% per<br />

patient-year. 2 The incidence is 0.5% to 6% in the aortic and/<br />

or mitral positions and up to 20% in the tricuspid position,<br />

whereas the risk of thrombosis in spite of adequate oral<br />

anticoagulation has been estimated at between 1% and<br />

4% per year. 3 Although inadequate anticoagulant therapy<br />

remains the main cause of this complication, it seems that<br />

lower pressures on the right side of the heart with a slower<br />

blood flow across the tricuspid valve is the most important<br />

cause of higher risk of thrombus formation in prosthetic<br />

tricuspid valves. 4, 5 In contrast to left-sided PVT, there is a<br />

paucity of data on the various aspects of tricuspid PVT. We<br />

herein present three cases of tricuspid PVT with different<br />

management approaches, namely thrombolytic therapy,<br />

conservative management, and re-replacement surgery, and<br />

then review the relevant literature.<br />

*<br />

Corresponding Author: Mohammad Javad Alemzadeh-Ansari, Department of Cardiology, Tehran University of Medical Sciences, Tehran Heart<br />

Center, Karegar Street, Tehran, Iran. 1411713138. Tel: +98 21 88029600. Fax: +98 21 88029731. Email: aansari@razi.tums.ac.ir.<br />

The Journal of Tehran University Heart Center 147


The Journal of Tehran University Heart Center<br />

Case Reports<br />

Case # 1<br />

A 32-year-old woman was admitted to our hospital with<br />

the complaint of dyspnea (The New York Heart Association<br />

[NYHA] factional class III) in January, 2004. Transthoracic<br />

echocardiography (TTE) showed left ventricular ejection<br />

fraction of 55%, severe mitral stenosis, mild mitral<br />

regurgitation, severe aortic regurgitation with moderate to<br />

severe aortic stenosis, and severe tricuspid regurgitation<br />

with moderate to severe tricuspid stenosis. In April 2004,<br />

the patient underwent three valves replacement surgery,<br />

during which she received a 24-mm St. Jude Valve (St. Jude<br />

Medical, Inc., St. Paul, MN, USA) in the mitral position,<br />

a 31-mm St. Jude Valve (St. Jude Medical, Inc., St. Paul,<br />

MN, USA) in the tricuspid position, and a 19-mm Regent<br />

Mechanical Prosthesis (St. Jude Medical, Inc., St. Paul, MN,<br />

USA) in the aortic position. Also, due to persistent atrial<br />

fibrillation with a slow ventricular response, she underwent<br />

permanent transvenous epicardial pacemaker placement<br />

during hospitalization. Twenty-one days after valvular<br />

surgery, TTE revealed that the function and gradients of the<br />

three prosthetic valves were within the acceptable range and<br />

the peak and mean gradients in the prosthetic tricuspid valve<br />

were 5 mm Hg and 3 mm Hg, respectively. At the time of<br />

discharge, the patient’s international normalized ratio (INR)<br />

was 3.9. She was discharged from the hospital with the<br />

recommendation to use Warfarin (with goal INR 2.5 - 3.5)<br />

plus 80 mg Aspirin daily.<br />

In February, 2006, the patient was re-admitted with the<br />

complaint of fatigue and palpitation. The pacemaker had a<br />

normal function. At the time of presentation, her INR was<br />

2.5. TTE revealed that the prosthetic aortic and mitral vales<br />

had normal functions and gradients, whereas the prosthetic<br />

tricuspid valve had malfunction with high gradients.<br />

Fluoroscopic evaluation revealed that there was no motion<br />

in both leaflets of the prosthetic tricuspid valve, while the<br />

motion of both other prosthetic valves was complete and<br />

within the normal range. With the diagnosis of tricuspid<br />

PVT, the patient was prescribed 250,000 U of Streptokinase<br />

via a peripheral vein over thirty minutes, followed by an<br />

intravenous infusion of 100,000 U per hour of Streptokinase<br />

for forty-eight hours. On the next day, fluoroscopic<br />

evaluation showed no evidence of prosthetic tricuspid valve<br />

malfunction, and the mobility of both leaflets was completely<br />

restored. At discharge, the patient’s INR was 2.6. She was<br />

discharged from the hospital with the recommendation to use<br />

Warfarin (with goal INR 3.0 - 3.5) plus 80 mg of Aspirin<br />

daily.<br />

In November, 2006, the patient was re-admitted to our<br />

hospital with the complaint of atypical chest pain. At<br />

presentation, her INR was 1.8 and her other laboratory<br />

data and also electrocardiogram were unremarkable. TTE<br />

Ahmad Yaminisharif et al.<br />

revealed that the prosthetic aortic and mitral vales had<br />

normal functions and gradients, whereas the prosthetic<br />

tricuspid valve had malfunction with high gradients.<br />

Fluoroscopic evaluation revealed that both leaflets of the<br />

prosthetic tricuspid valve were fixed without any motion,<br />

whereas the motion of both other prosthetic valves was<br />

complete and within the normal range. With the diagnosis<br />

of recurrent tricuspid PVT, the patient was administered a<br />

total of 35 mg of Tenecteplase: 15 mg bolus, followed by<br />

20 mg for four hours (the patient’s weight was 64 kg). On<br />

the next day, fluoroscopic evaluation showed no evidence<br />

of prosthetic tricuspid valve malfunction, and the mobility<br />

of both leaflets was completely restored. Twelve days after<br />

the administration of Tenecteplase, TTE demonstrated that<br />

a significant reduction had occurred in the gradient across<br />

the prosthetic tricuspid valve (9.5 mm Hg peak gradient,<br />

4.9 mm Hg mean gradient). At discharge, the patient’s INR<br />

was 3.5. She was discharged from the hospital with the<br />

recommendation to use Warfarin (with goal INR 3.0 - 3.5)<br />

plus 80 mg of Aspirin daily.<br />

Because of the battery depletion of the pacemaker, the<br />

patient was re-admitted for generator replacement on October<br />

30, 2011. She had discontinued Aspirin two years previously<br />

due to gastrointestinal problems. She complained of atypical<br />

chest pain of two months’ duration, but her myocardial<br />

perfusion scan was normal. TTE showed left ventricular<br />

ejection fraction of 55%, severe increased gradient in the<br />

prosthetic tricuspid valve with no paravalvular leakage, and<br />

normal function of the mitral and aortic prosthetic valves.<br />

Fluoroscopic evaluation of the prosthetic tricuspid valve<br />

revealed a severe drop in the motion of both leaflets. The<br />

patient was candidated for thrombolytic therapy with the<br />

diagnosis of recurrent tricuspid PVT. A single dose of 35<br />

mg of Tenecteplase was administrated via a peripheral vein,<br />

according to the dosing regimen used for acute myocardial<br />

infarction (the patient’s weight was 65 kg). Fluoroscopic<br />

evaluation exhibited no evidence of prosthetic tricuspid<br />

valve malfunction, and the mobility of both leaflets was<br />

completely restored. Seven days later, TTE demonstrated that<br />

a significant reduction had occurred in the gradients across<br />

the prosthetic tricuspid valve (9 mm Hg peak gradient, 4 mm<br />

Hg mean gradient). She was discharged from the hospital<br />

with the recommendation to use Warfarin (with goal INR<br />

3.0 - 3.5) plus 80 mg of Aspirin daily. Table 1 illustrates the<br />

three episodes of tricuspid PVT in our patient and the three<br />

successful methods for thrombolytic therapy.<br />

Case # 2<br />

A 49-year-old woman was admitted to our hospital for the<br />

replacement of the generator of a pacemaker in May 2011.<br />

Thirteen years previously in another center, because of severe<br />

aortic regurgitation and severe tricuspid regurgitation, she<br />

148


Prosthetic Tricuspid Valve Thrombosis: Three Case Reports and Literature Review<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Table 1. Three episodes of tricuspid PVT in case 1 and successful thrombolytic therapy<br />

Episode<br />

Time from valve<br />

replacement<br />

TTE findings in prosthetic tricuspid valve<br />

(before thrombolytic therapy) Thrombolytic drug Dosage<br />

Peak gradient<br />

Mean gradient<br />

1 22 months 14 mm Hg 11 mm Hg Streptokinase<br />

2 31 months 22 mm Hg 10 mm Hg Tenecteplase<br />

250,000 U loading dose followed<br />

by 100,000 U/h for 48 hours<br />

15 mg bolus, followed by 20 mg<br />

for 4 hours<br />

3 93 months 17 mm Hg 8.5 mm Hg Tenecteplase 35 mg bolus<br />

PVT, Prosthetic valve thrombosis; TTE, Transthoracic echocardiogram<br />

had undergone two valves replacement surgery, during which<br />

she received a 31-mm St. Jude Valve (St. Jude Medical, Inc.,<br />

St. Paul, MN, USA) in the tricuspid position and a 21-mm St.<br />

Jude Valve (St. Jude Medical, Inc., St. Paul, MN, USA) in the<br />

aortic position. After surgery, due to the presence of complete<br />

heart block, she underwent permanent epicardial pacemaker<br />

placement during hospitalization. But during this period, she<br />

was admitted frequently because of infection at the site of the<br />

pacemaker. At the time of recent presentation, her INR was<br />

1.9. TTE revealed that the prosthetic aortic valve had normal<br />

function and gradients, whereas the prosthetic tricuspid<br />

valve had malfunction with high gradients (15 mm Hg peak<br />

gradient, 10 mm Hg mean gradient). Fluoroscopic evaluation<br />

revealed that the motion of one leaflet of the prosthetic<br />

tricuspid valve was very restricted, while the other leaflet<br />

was fixed in an open position. The patient was symptomless.<br />

On physical examination, except for a holosystolic murmur<br />

at the left lower sternal border, there were no signs of heart<br />

failure. With the diagnosis of tricuspid PVT, the patient was<br />

administered 250,000 U of Streptokinase via a peripheral<br />

vein over thirty minutes, followed by an intravenous infusion<br />

of 100,000 U per hour of Streptokinase for forty-eight<br />

hours. Fluoroscopic evaluation showed no improvement<br />

in the motion of the leaflets. The patient was candidated<br />

for tricuspid valve replacement, but because of fungal<br />

infection in the pacemaker pocket and lead, replacement of<br />

the prosthetic valve or the generator was not performed. She<br />

was discharged from the hospital with the recommendation<br />

to use antibiotics with Warfarin (with goal INR 3.0 - 3.5)<br />

plus 80 mg of Aspirin daily. Six months later, she referred<br />

to our clinic; she had no signs or symptoms of heart failure.<br />

She was re-admitted for the replacement of the generator of<br />

the pacemaker. Before replacement, fluoroscopic evaluation<br />

showed no evidence of improvement in the motion of the<br />

prosthetic tricuspid valve leaflets. Because the patient was<br />

symptom-free, did not respond to thrombolytic therapy, and<br />

had a high risk for infection after valve replacement, surgery<br />

for valve replacement was not performed. After generator<br />

replacement, she was discharged with the recommendation<br />

to use Warfarin (with goal INR 3.0 - 3.5) plus 80 mg of<br />

Aspirin daily.<br />

Case # 3<br />

A 28-year-old woman was admitted to our hospital with the<br />

complaint of orthopnea and dyspnea (NYHA factional class<br />

III) in September, 2008. The electrocardiogram revealed<br />

atrial fibrillation. TTE showed left ventricular ejection<br />

fraction of 40%, severe mitral stenosis, severe tricuspid<br />

regurgitation, and a large mobile clot in the left atrium (2.7<br />

× 2.0 cm). She underwent two valves replacement surgery,<br />

during which she received a 31-mm St. Jude Valve (St.<br />

Jude Medical, Inc., St. Paul, MN, USA) in the tricuspid<br />

position and a 29-mm St. Jude Valve (St. Jude Medical,<br />

Inc., St. Paul, MN, USA) in the mitral position. Also in the<br />

same section of surgery, TEE revealed a large mobile clot<br />

in the left atrium without any lesion in the other chambers;<br />

the large clot was removed. After surgery, she received an<br />

intravenous bolus dose of 5000 U of heparin, followed by<br />

intravenous heparin (20000 U per day in divided doses).<br />

Because of persistent atrial fibrillation with a low ventricular<br />

response after surgery, eleven days later, she underwent<br />

permanent transvenous epicardial pacemaker placement.<br />

However, during implantation, fluoroscopy revealed that<br />

the motion of one leaflet of the prosthetic tricuspid valve<br />

was very restricted. With the diagnosis of the malfunction<br />

of the prosthetic tricuspid valve, she was transferred to the<br />

operating room again. During the removal of the previous<br />

prosthetic tricuspid valve, a large clot was seen in the<br />

right atrium which was attached to the leaflet. Thus, all of<br />

the thrombus was removed and a new 29-mm Hancock II<br />

bioprosthesis (Medtronic Inc., Minneapolis, Minn.) was<br />

placed in the tricuspid position. Because of the displacement<br />

of the transvenous epicardial lead during surgery, a new<br />

epicardial lead was placed. After surgery, TEE demonstrated<br />

an acceptable gradient across the tricuspid valve (5 mm Hg<br />

peak gradient, 2 mm Hg mean gradient). She was discharged<br />

with the recommendation to use Warfarin (with goal INR 3.0<br />

- 3.5) plus 80 mg of Aspirin daily.<br />

The Journal of Tehran University Heart Center149


The Journal of Tehran University Heart Center<br />

Discussion<br />

Pathogenesis<br />

Many studies have demonstrated that the leading cause<br />

of PVT is subtherapeutic anticoagulation, which chimes<br />

in with the findings in our two cases. 6-10 This is most often<br />

due to either patient noncompliance or iatrogenic cessation<br />

of anticoagulants in preparation for another procedure.<br />

Furthermore, valve design and materials influence the<br />

incidence of thrombotic complications. Some mechanisms<br />

have a role in PVT formation such as molecular interactions<br />

and influence of transprosthetic blood flow. Molecular<br />

interaction occurs between corpuscular blood components,<br />

plasma, and artificial surfaces. The initial adsorption of<br />

plasma proteins (fibrinogen, fibronectin, von Willebrand<br />

factor, vitronectin, and thrombospondin) on the artificial<br />

surface is generally followed by platelet adhesion. The<br />

passage of blood through the prosthetic valve creates a<br />

turbulent flow with shear stress, which gives rise to a<br />

structurally and metabolically damaged endocardium and<br />

thus reduces its resistance to thrombosis. Also, subclinical<br />

hemolysis with the release of adenosine diphosphatase,<br />

platelet factor 4, beta-thromboglobulin, and other proteins<br />

triggers the activation of the plasma coagulation system.<br />

Other intrinsic factors can progress to thrombus formation;<br />

these factors include loss of active atrial contractions (atrial<br />

fibrillation), presence of some systemic diseases (e.g.<br />

systemic lupus erythematosus) or malignant tumors, and<br />

incomplete endothelization of the sewing ring. Use of some<br />

drugs such as contraceptives leads to hypercoagulability<br />

state 3, 11-14 Recently, Ricome et al. reported two cases of PVT<br />

secondary to heparin-induced thrombocytopenia. 15<br />

Type and position of the prosthetic valve and time from<br />

1, 13, 16, 17<br />

surgery can influence thrombus formation (Table 2).<br />

Additionally, some studies have indicated that season can be<br />

correlated with an increased risk for thrombotic events. 18-20<br />

Piper et al. reported that PVT during winter months occurred<br />

more frequently than in the other seasons. 13<br />

Signs and Symptoms<br />

In contrast to the acute presentation of left-sided PVT, the<br />

onset of the symptoms of tricuspid PVT is usually insidious,<br />

and its diagnosis is often delayed. Sometimes symptoms<br />

are so slight that the patient is likely to have suffered from<br />

them for months or even a year without feeling the need for<br />

referral to the hospital. 4 Sometimes, the patient even may<br />

have no symptoms related to the tricuspid vale thrombosis,<br />

and the thrombosis is detected only during routine clinical<br />

examination. 4, 6 However, the involvement of both leaflets is<br />

usually required to produce symptoms. 6 The most frequent<br />

symptoms related to tricuspid valve malfunction include<br />

4, 6,<br />

dyspnea, ascites, peripheral edema, and systemic emboli.<br />

21, 22<br />

Also, sometimes the disappearance or attenuation of the<br />

prosthetic valve noise may be reported by the patient and/<br />

or relatives. Moreover, in some cases where an interatrial<br />

communication is present, a pulmonary embolus or a leftsided<br />

embolic event may be the presenting manifestation of<br />

23, 24<br />

tricuspid PVT.<br />

It seems that physical examination may provide important<br />

clues for the diagnosis of tricuspid PVT, compared to the<br />

thrombosis of the left side, which normally has more severe<br />

symptoms. 25 Absence or muffling of prosthetic sounds<br />

in the tricuspid position might be noted. Other findings<br />

include auscultation of a new holosystolic murmur located<br />

at the left lower sternal border or in the subxiphoid region<br />

that may increase with inspiration or maneuvers that<br />

increase venous return (denoting the presence of tricuspid<br />

regurgitation), auscultation of a new murmur that is low in<br />

frequency, diastolic, located at the lower left sternal border<br />

or infraxiphoid area, and increases with inspiration and other<br />

maneuvers that increase tricuspid flow velocity (denoting<br />

tricuspid stenosis), or auscultation of a combination<br />

of murmurs that characterize both of these conditions.<br />

Sometimes mid-diastolic and/or pan-systolic murmurs can<br />

be heard in the tricuspid area (Zhang DY, Lozier J, Chang R,<br />

Sachdev V, Chen MY, Audibert JL, Horvath KA, Rosing DR.<br />

Case study and review: Treatment of tricuspid prosthetic<br />

valve thrombosis. Int J Cardiol 2011 Oct 14. [Epub ahead<br />

of print]).<br />

Diagnosis<br />

Ahmad Yaminisharif et al.<br />

The onset of the symptoms of tricuspid PVT is usually<br />

insidious and sometimes the patient has nonspecific<br />

symptoms or is even symptomless; therefore, suspicion of<br />

tricuspid PVT may be raised by physical findings, symptoms<br />

of heart failure, or rarely the diagnosis of embolization,<br />

especially in patients with poor anticoagulation therapy 8,<br />

17, 25<br />

NYHA has classified PVT in functional classes I to<br />

IV. The non-obstructive forms of PVT (NYHA functional<br />

Table 2. Thrombogenicity of mechanical prosthetic valve based on type, position, and time from surgery<br />

Lower<br />

Higher<br />

Valve type Bileaflet-tilting-disk Caged-ball single-tilting-disk<br />

Position Mitral or aortic Tricuspid<br />

Time from replacement After 3 months First 3 months<br />

150


Prosthetic Tricuspid Valve Thrombosis: Three Case Reports and Literature Review<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

classes I or II) are usually incidental echocardiographic<br />

findings in patients with symptomless, nonspecific<br />

symptoms, or thromboembolic events. The obstructive<br />

forms (NYHA functional classes III or IV) correspond to<br />

obvious hemodynamic repercussions, sometimes including<br />

cardiogenic shock, and are often associated with cerebral or<br />

3, 11, 26<br />

peripheral embolism.<br />

Imaging modalities in patients with suspected PVT are<br />

aimed to evaluate leaflet immobilization, cause of leaflet<br />

immobilization (thrombus versus pannus or both), and<br />

whether thrombolytic therapy attempt in the patient would<br />

be successful. Usually the first modality for detecting<br />

prosthetic tricuspid valve dysfunction is TTE. Montorsi<br />

et al. reported that sensitivity, specificity, and positive and<br />

negative predictive values for the diagnosis of PVT in the<br />

mitral or aortic position by TTE were 75%, 64%, 57%, and<br />

78%, respectively. TTE, especially in experienced hands, can<br />

detect increased transvalvular gradients (mean of 6 mm Hg<br />

or higher, and peak of 15 mm Hg or higher), pressure halftime<br />

of 230 msec or higher, transvalvular gradients of 50%<br />

or higher than that observed before, wide intravalvular jet of<br />

tricuspid regurgitation, lower orifice area, visible thrombus<br />

on the prosthetic valve, and inability to demonstrate two<br />

different mobile echoes representing the valve leaflets<br />

in a high quality image. Indirect, nonspecific signs are an<br />

3, 6, 27<br />

enlarged right atrium and engorged inferior vena cava.<br />

After performance of TTE, the diagnosis should be<br />

confirmed by more specific modalities, namely fluoroscopy<br />

or TEE. 28 Fluoroscopy is a non-invasive method for<br />

detecting PVT, especially in patients with bileaflet prosthetic<br />

valves, and have high clinical suspicious for PVT and<br />

normal Doppler study. 29 Sensitivity, specificity, and positive<br />

and negative predictive values for the diagnosis of PVT in<br />

the mitral or aortic position by fluoroscopy are 87%, 78%,<br />

80%, and 91%. 28 Also, fluoroscopy has an important role for<br />

detecting the response to thrombolytic therapy. Thrombolysis<br />

significantly reduces the mean pressure gradient and<br />

improves valve leaflet opening angle. But some patients<br />

whose pressure gradient normalizes after thrombolytic<br />

infusion tend to continue to have concomitant abnormal<br />

leaflet motion at fluoroscopy, suggesting incomplete<br />

resolution of valve obstruction (pseudo responders). If lytic<br />

infusion is stopped at this time, the remaining thrombus<br />

could be the trigger for a late rethrombotic process. Thus,<br />

fluoroscopy should be carried out at regular intervals during<br />

therapy to confirm Doppler changes. 29<br />

TEE can correctly identify opening and closing angles in<br />

all patients, regardless of the prosthetic type. 30 TEE should<br />

be performed in selected patients even if fluoroscopy is<br />

negative because TEE is an invasive modality. On the other<br />

hand, fluoroscopy and TTE can correctly identify PVT in<br />

85% of all cases. Thus, fluoroscopy and TTE are quick,<br />

effective, and complementary diagnostic tools for the<br />

diagnosis of PVT in most patients. 28 Despite the scarcity of<br />

data on the role of TEE in diagnosing tricuspid PVT, it seems<br />

that if there is high clinical suspicion and other diagnostic<br />

modalities are not helpful, TEE will be help. 27 Furthermore,<br />

TEE is a superior modality for detecting the etiology of the<br />

obstruction (thrombus versus pannus), size, and location of<br />

the thrombus compared with TTE and fluoroscopy. 31<br />

Magnetic Resonance Imaging (MRI) and cardiac<br />

catheterization have limited diagnostic roles, because TEE and<br />

fluoroscopy can provide adequate data for decision-making.<br />

Since MRI is more expensive and time-consuming than<br />

echocardiography, it should be used only when prostheticvalve<br />

regurgitation or paravalvular leakage is suspected but<br />

not adequately visualized by echocardiography. 1 In contrast,<br />

Cardiac Multi-Detector Computer Tomography can provide<br />

sharp images to characterize quantitatively the reduced<br />

mobility of prosthetic leaflets or even directly visualize and<br />

distinguish between thrombus and pannus. 32-34<br />

Treatment<br />

There are different therapeutic modalities available for<br />

PVT such as heparin treatment, thrombolysis, surgery, or<br />

even in some cases only watchful waiting. Selecting one<br />

of these modalities is largely influenced by the presence of<br />

valvular obstruction, valve location (left- or right-sided), and<br />

clinical status. 6, 35 Surgery is more frequently performed for<br />

the treatment of left-sided PVT, not least in patients with<br />

either NYHA functional class III–IV symptoms or a large<br />

clot burden 36 and thrombolytic therapy is more favorable for<br />

right-sided PVT, because the risk of systemic embolization<br />

and recurrence rate is high by thrombolytic therapy in left-<br />

37, 38<br />

sided PVT.<br />

The conservative continued anticoagulation approach<br />

in patients with tricuspid PVT would only be appropriate<br />

if there is no significant hemodynamic compromise or<br />

a contraindication to either surgery or pharmacologic<br />

intervention is present. Shapira et al. reported that<br />

asymptomatic patients with tricuspid PVT who did not<br />

respond to thrombolytic therapy might be discharged from<br />

the hospital with long-term intensified anticoagulant therapy<br />

and close follow-up. The leaflet motion can be fully restored<br />

later. 6 However, Montorsi et al. proposed that leaflet mobility<br />

and duration of prosthetic valve symptoms were important<br />

factors in determining successful thrombolytic therapy. It<br />

may be because the amount of the thrombus that led to the<br />

stuck valve was minimal, thereby improving the chance of<br />

successful thrombolytic therapy. 29<br />

According the guidelines of the American Heart Association<br />

/American College of Cardiology (AHA/ACC) and the<br />

American College of Chest physicians (ACCP), in contrast to<br />

left-sided PVT, thrombolytic therapy is reasonable for rightsided<br />

PVT with NYHA functional class III-IV symptoms or<br />

a large clot burden. 36, 39 It is due to the high success rate and<br />

The Journal of Tehran University Heart Center151


The Journal of Tehran University Heart Center<br />

Ahmad Yaminisharif et al.<br />

low incidence of embolism compared to left-sided PVT. The<br />

European Society of Cardiology guideline also recommends<br />

thrombolytic therapy for tricuspid PVT, but this guideline<br />

does not mention the NYHA functional class or clot burden. 40<br />

Long-standing symptoms even more than a month should<br />

not make one reluctant to use thrombolytic therapy. 6 If<br />

thrombolytic therapy fails, the presence of a large thrombus<br />

or pannus should be considered, which may require surgical<br />

intervention (thrombectomy or valve replacement).<br />

In contrast to left-sided PVT, there is limited information<br />

about thrombolytic therapy for tricuspid PVT. Various<br />

thrombolytic agents have been used for PVT, including<br />

streptokinase, urokinases, and tissue-type plasminogen<br />

activator (tPA). The choice of the thrombolytic agent depends<br />

on several factors, including cost, time to attain maximal<br />

pharmacologic effect, half-life of the thrombolytic agent,<br />

and hemorrhagic complications. Amongst the above agents,<br />

streptokinase is cheaper and has lower cerebral hemorrhage<br />

rates. In contrast, tPA has a faster effect reversion and<br />

faster reach to maximal pharmacologic effect. 3 Roudaut<br />

et al. indicated that patients treated by streptokinase had a<br />

significantly full success rate compared to patients treated<br />

by tPA or urokinases (86%, 68%, and 59%, respectively).<br />

Nonetheless, combined therapy improved the results of<br />

thrombolytic therapy in all the groups. 41 Also, they concluded<br />

that full success by thrombolytic therapy was higher in<br />

patients in NYHA functional classes I or II; nevertheless,<br />

they did not find a significant difference between patients<br />

with tilting-discs and bileaflet valves, or between patients<br />

with first episode of thrombosis and recurrent thrombotic<br />

episodes groups. 41 The dosage and route of the administration<br />

of thrombolytic therapy are different in various studies.<br />

Hering et al. recommended using streptokinase by starting<br />

a bolus dose of 250,000 IU over thirty minutes, followed<br />

by an intravenous infusion of 100,000 IU per hour (same<br />

as the therapy of our patient in the first episode of PVT),<br />

urokinase by the same protocol used in patients with acute<br />

pulmonary embolism, and t-PA at a dosage of 100 mg given<br />

over a period of two to five hours. 14 Caceres-Loriga et al.<br />

recommended using streptokinase by starting a bolus dose of<br />

250,000 IU over three minutes, followed by an intravenous<br />

infusion of 100,000 IU per hour (maximum duration of<br />

seventy-two hours), urokinase by starting a bolus dose of<br />

4500 U/kg, followed by an intravenous infusion of 4500<br />

U/kg/h (maximum duration of twenty-four to forty-eight<br />

hours), and tPA by starting a bolus dose of 15 mg over five<br />

minutes, followed by an intravenous infusion of 95 mg over<br />

ninety minutes. 3 Manteiga et al. used the short-course of<br />

thrombolytic therapy as a first line for PVT: streptokinase<br />

by starting a bolus dose of 250,000 IU over twenty minutes,<br />

followed by an intravenous infusion of 1,500,000 IU over<br />

ninety minutes, or tPA by starting a bolus dose of 10 mg,<br />

followed by an intravenous infusion of 90 mg over ninety<br />

minutes. They concluded that a successful rate by these<br />

regimes was 82%. 42 However, Alpert recommended another<br />

dose for streptokinase in right-sided PVT (starting a dose of<br />

500,000 IU over twenty minutes, followed by an intravenous<br />

infusion of 1,500,000 IU over ninety minutes). 43 Some<br />

other investigators have used direct intra-atrial infusion of<br />

thrombolytic for PVT. 44, 45 For the first time, Zhang et al.<br />

reported a case of tricuspid PVT, which was successfully<br />

treated by an intra-right atrium infusion of tPA.<br />

Recently, Tenecteplase (a genetically engineered variant<br />

of tPA which has a longer half-life than tPA and is resistant<br />

to inactivation by plasminogen activator inhibitor-1 46 ) has<br />

been utilized for PVT. Our literature search shows that<br />

Tenecteplase has been used in limited case repots for mitral<br />

or aortic PVT. 47-52 Although Tenecteplase has been prescribed<br />

in different doses and via different methods, we used this<br />

thrombolytic according to the dosing regimen employed<br />

for acute myocardial infarction. Data on Tenecteplase for<br />

the treatment of PVT are limited. Still, Melandri et al. in a<br />

review study about patients with acute myocardial infarction<br />

showed that this drug had some advantages compared with<br />

tPA. These advantages included being more fibrin-specific,<br />

usability in a single bolus dose, and having less non-cerebral<br />

bleeding. Be that as it may, mortality rates and intracranial<br />

hemorrhage rates were similar to those of tPA. 46 For the first<br />

time, we reported a successful use of Tenecteplase in our<br />

case 1 for the treatment of two episodes of recurrent tricuspid<br />

PVT. It seems that this drug might be a suitable alternative<br />

for the other types of tPA in the future.<br />

If thrombolytic therapy is successful, a continuous<br />

infusion of unfractionated heparin is indicated and should<br />

be initiated. Moreover, activated partial thromboplastin<br />

time should be maintained at twofold the baseline values,<br />

followed by conversion to oral anticoagulation combined<br />

with Aspirin (50 to 100 mg per day). 39 In contrast to leftsided<br />

PVT, guidelines do not provide a recommended INR<br />

for prostheses in the tricuspid position. 36, 39, 40 For bileaflet<br />

prosthetic valves in the mitral position, a range of 2.5 - 3.5<br />

is recommended, 2.0 - 3.0 in the aortic position for patients<br />

without additional risk factors for thromboembolism. 39<br />

However, Shapira et al. recommended target INR levels of<br />

3.5 - 4.0 for patients with tricuspid prostheses 6 and Zhang<br />

et al., in order to prevent future thrombotic complications<br />

of tricuspid PVT, considered target INR levels of 3.0 - 3.5.<br />

Recurrent tricuspid PVT<br />

A major disadvantage of thrombolytic therapy is the<br />

relatively high incidence of recurrent thrombosis during<br />

follow-up; however, data are limited about rethrombosis<br />

after thrombolytic therapy of tricuspid PVT. Recurrent<br />

rates after thrombolytic therapy vary from 11% to 31%. 3,<br />

53-58<br />

Overall, risk of recurrent thrombosis after thrombolytic<br />

therapy in left-sided prosthetic valves is higher than that in<br />

152


Prosthetic Tricuspid Valve Thrombosis: Three Case Reports and Literature Review<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

the tricuspid position. A meta-analysis showed that while<br />

the incidence of recurrent thrombosis in left-sided prosthetic<br />

valves was 20%, it was 14% in the tricuspid position. 14 As<br />

was mentioned earlier, the etiology of recurrent thrombosis<br />

is the same as that of the first episode. Also, the coexistence<br />

of thrombus and pannus tissue on a prosthetic valve is<br />

another factor that can explain the recurrence of PVT after<br />

successful thrombolytic therapy in a subset of patients. 53,<br />

56<br />

However, some recurrences may be the result of an<br />

uncompleted resolution of the initial thrombotic process<br />

rather than the result of a new thrombosis. Thus, after<br />

successful thrombolytic therapy, it is very important to<br />

follow up patients with serial clinical and echocardiographic<br />

examinations. 27 The results of rethrombolysis after PVT<br />

recurrence are comparable to those obtained after the first<br />

thrombolytic therapy, which is concordant with our case 1. 38,<br />

41, 58-62<br />

Therefore, rethrombolysis is safe with a high successful<br />

rate and is recommended in patients with recurrent tricuspid<br />

PVT.<br />

Choose a prosthetic valve: mechanical or biological<br />

Tricuspid valve replacement is one of the most challenging<br />

operations of cardiac surgery. Most cardiac surgeons<br />

undertake tricuspid valve surgery infrequently and usually<br />

perform tricuspid valve repair. Incidence of tricuspid<br />

valve replacement is approximately 0.7% of all valve<br />

replacements. 63 Although many studies have been performed<br />

to determine the preference between mechanical or biological<br />

valves in the tricuspid position, they have not reached the<br />

same conclusion yet. 63-69 As was mentioned before, because<br />

the risk of thrombosis is high in the tricuspid position and<br />

thrombus formation is lower in biological valves, 65, 66, 70 we<br />

recommend the use of biological valves in the tricuspid<br />

position, which is similar to that in re-replacement valve<br />

surgery (the same as our case 3).<br />

Conclusion<br />

Thrombosis in tricuspid prosthetic valves is high and<br />

in some cases, patients are symptom-free or have a mild<br />

complaint. Thus, regular visits after vale replacement are<br />

reasonable and if there is suspicion of PVT, other modalities<br />

(first TTE) are recommended. Herein, we reported three cases<br />

of tricuspid PVT with different conditions. The main cause<br />

of PVT in our cases was subtherapeutic anticoagulation. The<br />

first case was a woman who suffered from recurrent PVT. In<br />

this case we successfully used for the first time Tenecteplase<br />

for second and third episodes. Given that this drug can be<br />

used in a single dose and has acceptable efficacy compared<br />

to the other conventional thrombolytic agents, we would<br />

recommend Tenecteplase as a good alternative for PVT<br />

treatment. Also, this case shows us that thrombolytic therapy<br />

is a good option for recurrent tricuspid PVT, in contrast to<br />

left-sided PVT. The second case had fixed leaflets in open<br />

position, while the patient was symptomless. Thrombolytic<br />

therapy failed in this case; however, due to the patient’s<br />

chronic infection, we could not replace her valve. At six<br />

months’ follow-up, the motion of the leaflets was restricted<br />

and she was symptom-free. Thus, if thrombolytic therapy<br />

fails, surgery is not possible, the patient is symptom-free,<br />

and hemodynamic is stable, close observation with oral<br />

anticoagulant would be a reasonable course of action. The<br />

last case was a woman who had a large thrombus in the<br />

right atrium immediately after mitral and tricuspid valve<br />

replacement. We think that the cause of thrombus formation<br />

in this case was inadequate anticoagulation therapy. The<br />

patient underwent re-replacement surgery and a new<br />

biological valve was implanted in the tricuspid position.<br />

References<br />

1. Butany J, Ahluwalia MS, Munroe C, Fayet C, Ahn C, Blit P, Kepron<br />

C, Cusimano RJ, Leask RL. Mechanical heart valve prostheses:<br />

identification and evaluation (erratum). Cardiovascular Pathology<br />

2003;12:322-344.<br />

2. Vongpatanasin W, Hillis LD, Lange RA. Prosthetic heart valves. N<br />

Engl J Med 1996;335:407-416.<br />

3. Caceres-Loriga FM, Perez-Lopez H, Santos-Gracia J, Morlans-<br />

Hernandez K. Prosthetic heart valve thrombosis: pathogenesis,<br />

diagnosis and management. Int J Cardiol 2006;110:1-6.<br />

4. Thorburn CW, Morgan JJ, Shanahan MX, Chang VP. Long-term<br />

results of tricuspid valve replacement and the problem of prosthetic<br />

valve thrombosis. Am J Cardiol 1983;51:1128-1132.<br />

5. Minami K, Horstkotte D, Schulte HD, Bircks W. Thrombosis of<br />

two St. Jude Medical prostheses in one patient after triple valve<br />

replacement. Case report and review of the literature. Eur J<br />

Cardiothorac Surg 1988;2:48-52.<br />

6. Shapira Y, Sagie A, Jortner R, Adler Y, Hirsch R. Thrombosis of<br />

bileaflet tricuspid valve prosthesis: clinical spectrum and the role<br />

of nonsurgical treatment. Am Heart J 1999;137:721-725.<br />

7. Buttard P, Bonnefoy E, Chevalier P, Marcaz PB, Robin J, Obadia<br />

JF, Kirkorian G, Touboul P. Mechanical cardiac valve thrombosis in<br />

patients in critical hemodynamic compromise. Eur J Cardiothorac<br />

Surg 1997;11:710-713.<br />

8. Lengyel M, Fuster V, Keltai M, Roudaut R, Schulte HD, Seward<br />

JB, Chesebro JH, Turpie AG. Guidelines for management of leftsided<br />

prosthetic valve thrombosis: a role for thrombolytic therapy.<br />

Consensus Conference on Prosthetic Valve Thrombosis. J Am Coll<br />

Cardiol 1997;30:1521-1526.<br />

9. Deviri E, Sareli P, Wisenbaugh T, Cronje SL. Obstruction of<br />

mechanical heart valve prostheses: clinical aspects and surgical<br />

management. J Am Coll Cardiol 1991;17:646-650.<br />

10. Kawano H, Oda T, Fukunaga S, Tayama E, Kawara T, Oryoji<br />

A, Aoyagi S. Tricuspid valve replacement with the St. Jude<br />

Medical valve: 19 years of experience. Eur J Cardiothorac Surg<br />

2000;18:565-569.<br />

11. Silber H, Khan SS, Matloff JM, Chaux A, DeRobertis M, Gray R.<br />

The St. Jude valve. Thrombolysis as the first line of therapy for<br />

cardiac valve thrombosis. Circulation 1993;87:30-37.<br />

12. Edmunds LH, Jr. Thrombotic and bleeding complications of<br />

prosthetic heart valves. Ann Thorac Surg 1987;44:430-445.<br />

13. Piper C, Hering D, Horstkotte D. Prosthetic valve thrombosis.<br />

predisposition and diagnosis. Eur Heart J Supplements 2001;3:Q16-21.<br />

The Journal of Tehran University Heart Center153


The Journal of Tehran University Heart Center<br />

Ahmad Yaminisharif et al.<br />

14. Hering D, Piper C, Horstkotte D. Management of prosthetic valve<br />

thrombosis. Eur Heart J Supplements 2001;3:Q22-26.<br />

15. Ricome S, Provenchere S, Aubier B, Ajzenberg N, Lepage L, Dilly<br />

MP, Dufour G, Montravers P, Longrois D. Two cases of valvular<br />

thrombosis secondary to heparin-induced thrombocytopenia<br />

managed without surgery. Circulation 2011;123:1355-1357.<br />

16. Cevik C, Izgi C, Dechyapirom W, Nugent K. Treatment of<br />

prosthetic valve thrombosis: rationale for a prospective randomized<br />

clinical trial. J Heart Valve Dis 2010;19:161-170.<br />

17. Pibarot P, Dumesnil JG. Prosthetic heart valves: selection of<br />

the optimal prosthesis and long-term management. Circulation<br />

2009;119:1034-1048.<br />

18. Wilhelmsen L, Svardsudd K, Korsan-Bengtsen K, Larsson B,<br />

Welin L, Tibblin G. Fibrinogen as a risk factor for stroke and<br />

myocardial infarction. N Engl J Med 1984;311:501-505.<br />

19. Kannel WB, Wolf PA, Castelli WP, D›Agostino RB. Fibrinogen<br />

and risk of cardiovascular disease. The Framingham Study. JAMA<br />

1987;258:1183-1186.<br />

20. Alderson M. Season and mortality. Health Trends 1985;17:87-96.<br />

21. Kao CL, Lu MS, Chang JP, Yang TY, Cheng HW. Thrombotic<br />

obstruction of a mechanical prosthetic valve in tricuspid position.<br />

Tex Heart Inst J 2009;36:261-263.<br />

22. Suwansirikul S, Glassman E, Raia F, Spencer FC. Late thrombosis<br />

of Starr-Edwards tricuspid ball valve prosthesis. Am J Cardiol<br />

1974;34:737-740.<br />

23. Oxenham H, Bloomfield P, Wheatley DJ, Lee RJ, Cunningham J,<br />

Prescott RJ. Twenty year comparison of a Bjork-Shiley mechanical<br />

heart valve with porcine bioprostheses. Heart 2003;89:715-721.<br />

24. Hammermeister K, Sethi GK, Henderson WG, Grover FL, Oprian<br />

C, Rahimtoola SH. Outcomes 15 years after valve replacement<br />

with a mechanical versus a bioprosthetic valve: final report<br />

of the Veterans Affairs randomized trial. J Am Coll Cardiol<br />

2000;36:1152-1158.<br />

25. Shapira Y, Vaturi M, Sagie A. Obstructive left-sided prosthetic<br />

valve thrombosis. Acute Card Care 2009;11:160-168.<br />

26. Reddy NK, Padmanabhan TN, Singh S, Kumar DN, Raju PR,<br />

Satyanarayana PV, Rao DP, Rajagopal P, Raju BS. Thrombolysis<br />

in left-sided prosthetic valve occlusion: immediate and follow-up<br />

results. Ann Thorac Surg 1994;58:462-470.<br />

27. Zoghbi WA, Chambers JB, Dumesnil JG, Foster E, Gottdiener<br />

JS, Grayburn PA, Khandheria BK, Levine RA, Marx GR, Miller<br />

FA, Jr, Nakatani S, Quiñones MA, Rakowski H, Rodriguez LL,<br />

Swaminathan M, Waggoner AD, Weissman NJ, Zabalgoitia<br />

M; American Society of Echocardiography›s Guidelines and<br />

Standards Committee; Task Force on Prosthetic Valves; American<br />

College of Cardiology Cardiovascular Imaging Committee;<br />

Cardiac Imaging Committee of the American Heart Association;<br />

European Association of Echocardiography; European Society<br />

of Cardiology; Japanese Society of Echocardiography; Canadian<br />

Society of Echocardiography; American College of Cardiology<br />

Foundation; American Heart Association; European Association<br />

of Echocardiography; European Society of Cardiology;<br />

Japanese Society of Echocardiography; Canadian Society of<br />

Echocardiography. Recommendations for evaluation of prosthetic<br />

valves with echocardiography and doppler ultrasound: a report<br />

From the American Society of Echocardiography›s Guidelines<br />

and Standards Committee and the Task Force on Prosthetic Valves,<br />

developed in conjunction with the American College of Cardiology<br />

Cardiovascular Imaging Committee, Cardiac Imaging Committee<br />

of the American Heart Association, the European Association of<br />

Echocardiography, a registered branch of the European Society<br />

of Cardiology, the Japanese Society of Echocardiography and the<br />

Canadian Society of Echocardiography, endorsed by the American<br />

College of Cardiology Foundation, American Heart Association,<br />

European Association of Echocardiography, a registered branch<br />

of the European Society of Cardiology, the Japanese Society of<br />

Echocardiography, and Canadian Society of Echocardiography. J<br />

Am Soc Echocardiogr 2009;22:975-1014.<br />

28. Montorsi P, De Bernardi F, Muratori M, Cavoretto D, Pepi M.<br />

Role of cine-fluoroscopy, transthoracic, and transesophageal<br />

echocardiography in patients with suspected prosthetic heart valve<br />

thrombosis. Am J Cardiol 2000;85:58-64.<br />

29. Montorsi P, Cavoretto D, Alimento M, Muratori M, Pepi M.<br />

Prosthetic mitral valve thrombosis: can fluoroscopy predict the<br />

efficacy of thrombolytic treatment Circulation 2003;108:II79-84.<br />

30. Muratori M, Montorsi P, Teruzzi G, Celeste F, Doria E, Alamanni<br />

F, Pepi M. Feasibility and diagnostic accuracy of quantitative<br />

assessment of mechanical prostheses leaflet motion by transthoracic<br />

and transesophageal echocardiography in suspected prosthetic<br />

valve dysfunction. Am J Cardiol 2006;97:94-100.<br />

31. Barbetseas J, Nagueh SF, Pitsavos C, Toutouzas PK, Quiñones MA,<br />

Zoghbi WA. Differentiating thrombus from pannus formation in<br />

obstructed mechanical prosthetic valves: an evaluation of clinical,<br />

transthoracic and transesophageal echocardiographic parameters. J<br />

Am Coll Cardiol 1998;32:1410-1417.<br />

32. Tsai IC, Lin YK, Chang Y, Fu YC, Wang CC, Hsieh SR, Wei HJ,<br />

Tsai HW, Jan SL, Wang KY, Chen MC, Chen CC. Correctness<br />

of multi-detector-row computed tomography for diagnosing<br />

mechanical prosthetic heart valve disorders using operative<br />

findings as a gold standard. Eur Radiol 2009;19:857-867.<br />

33. Teshima H, Hayashida N, Fukunaga S, Tayama E, Kawara T,<br />

Aoyagi S, Uchida M. Usefulness of a multidetector-row computed<br />

tomography scanner for detecting pannus formation. Ann Thorac<br />

Surg 2004;77:523-526.<br />

34. Habets J, Symersky P, van Herwerden LA, de Mol BA, Spijkerboer<br />

AM, Mali WP, Budde RP. Prosthetic heart valve assessment with<br />

multidetector-row CT: imaging characteristics of 91 valves in 83<br />

patients. Eur Radiol 2011;21:1390-1396.<br />

35. Roudaut R, Serri K, Lafitte S. Thrombosis of prosthetic heart valves:<br />

diagnosis and therapeutic considerations. Heart 2007;93:137-142.<br />

36. Bonow RO, Carabello BA, Chatterjee K, de Leon AC, Jr, Faxon<br />

DP, Freed MD, Gaasch WH, Lytle BW, Nishimura RA, O›Gara<br />

PT, O›Rourke RA, Otto CM, Shah PM, Shanewise JS; American<br />

College of Cardiology/American Heart Association Task Force<br />

on Practice Guidelines. 2008 focused update incorporated into<br />

the ACC/AHA 2006 guidelines for the management of patients<br />

with valvular heart disease: a report of the American College of<br />

Cardiology/American Heart Association Task Force on Practice<br />

Guidelines (Writing Committee to revise the 1998 guidelines for<br />

the management of patients with valvular heart disease). Endorsed<br />

by the Society of Cardiovascular Anesthesiologists, Society for<br />

Cardiovascular Angiography and Interventions, and Society of<br />

Thoracic Surgeons. J Am Coll Cardiol 2008;52:e1-142.<br />

37. Ozkan M, Kaymaz C, Kirma C, Sönmez K, Ozdemir N, Balkanay<br />

M, Yakut C, Deligönül U. Intravenous thrombolytic treatment<br />

of mechanical prosthetic valve thrombosis: a study using<br />

serial transesophageal echocardiography. J Am Coll Cardiol<br />

2000;35:1881-1819.<br />

38. Lengyel M, Horstkotte D, Völler H, Mistiaen WP; Working Group<br />

Infection, Thrombosis, Embolism and Bleeding of the Society for<br />

Heart Valve Disease. Recommendations for the management of<br />

prosthetic valve thrombosis. J Heart Valve Dis 2005;14:567-575.<br />

39. Salem DN, O›Gara PT, Madias C, Pauker SG; American College of<br />

Chest Physicians. Valvular and structural heart disease: American<br />

College of Chest Physicians Evidence-Based Clinical Practice<br />

Guidelines (8th Edition). Chest 2008;133:593S-629S.<br />

40. Vahanian A, Baumgartner H, Bax J, Butchart E, Dion R, Filippatos<br />

G, Flachskampf F, Hall R, Iung B, Kasprzak J, Nataf P, Tornos P,<br />

Torracca L, Wenink A; Task Force on the Management of Valvular<br />

Hearth Disease of the European Society of Cardiology; ESC<br />

Committee for Practice Guidelines. Guidelines on the management<br />

of valvular heart disease: The Task Force on the Management of<br />

Valvular Heart Disease of the European Society of Cardiology. Eur<br />

Heart J 2007;28:230-268.<br />

41. Roudaut R, Lafitte S, Roudaut MF, Courtault C, Perron JM, Jaïs<br />

C, Pillois X, Coste P, DeMaria A. Fibrinolysis of mechanical<br />

prosthetic valve thrombosis: a single-center study of 127 cases. J<br />

Am Coll Cardiol 2003;41:653-658.<br />

154


Prosthetic Tricuspid Valve Thrombosis: Three Case Reports and Literature Review<br />

42. Manteiga R, Carlos Souto J, Altès A, Mateo J, Arís A, Dominguez<br />

JM, Borrás X, Carreras F, Fontcuberta J. Short-course thrombolysis<br />

as the first line of therapy for cardiac valve thrombosis. J Thorac<br />

Cardiovasc Surg 1998;115:780-784.<br />

43. Alpert JS. The thrombosed prosthetic valve: current<br />

recommendations based on evidence from the literature. J Am Coll<br />

Cardiol 2003;41:659-660.<br />

44. Desai S, Kavinsky C. Localized left atrial administration of tPA<br />

for the treatment of mechanical mitral valve thrombosis. Catheter<br />

Cardiovasc Interv 2008;72:151-155.<br />

45. Seltzer SM, Reed MD, Siwik ES. Intra-atrial tissue plasminogen<br />

activator infusion for prosthetic valve thrombosis. Catheter<br />

Cardiovasc Interv 2006;67:139-141.<br />

46. Melandri G, Vagnarelli F, Calabrese D, Semprini F, Nanni S,<br />

Branzi A. Review of tenecteplase (TNKase) in the treatment of<br />

acute myocardial infarction. Vasc Health Risk Manag 2009;5:249-<br />

256.<br />

47. Maegdefessel L, Issa H, Scheler C, Thäle V, Schlitt A, Hartelt U,<br />

Grabitz R, Buerke M. 27-year old pregnant woman with syncope<br />

and dyspnea after aortic alloplastic heart valve replacement 15<br />

years ago. Internist (Berl) 2008;49:868-872.<br />

48. Charokopos N, Antonitsis P, Artemiou P, Rouska E, Foroulis<br />

C, Papakonstantinou C. Acute mechanical prosthetic valve<br />

thrombosis after initiating oral anticoagulation therapy. Is bridging<br />

anticoagulation with heparin required Interact Cardiovasc Thorac<br />

Surg 2009;9:685-687.<br />

49. Ferreiro-Gutierrez JL, riza-Sole A, Manas-Jimenez P, Ruiz-Majoral<br />

A. Repeated thrombolysis with tenecteplase as a bridge to valvular<br />

replacement in a case of preoclusive mitral prosthetic thrombosis.<br />

Med Clin (Barc) 2009;133:402-403.<br />

50. Al-Sarraf N, Al-Shammari F, Al-Fadhli J, Al-Shawaf E. Successful<br />

thrombolysis of a thrombosed prosthetic mitral valve using a<br />

synthetic tissue plasminogen activator: a case report. J Med Case<br />

Rep 2010;4:241.<br />

51. Slaoui M, Cherradi R, Ounzar M, Massou S, Srairi JE. Thrombosis<br />

valvular prosthesis of Starr treated successfully by tenecteplase<br />

during pregnancy. Ann Fr Anesth Reanim 2010;29:500-501.<br />

52. Ayyub Ghori M, Bakir S, Ellahham S, Al Nassir A, Al Zubaidi A,<br />

Augustin N, Ayman Abdelaziz M, Patrick Turrin N, Al Mahmeed<br />

WA. Tenecteplase in prosthetic mitral valve thrombosis. J Saudi<br />

Heart Assoc 2011;23:93-95.<br />

53. Keuleers S, Herijgers P, Herregods MC, Budts W, Dubois C,<br />

Meuris B, Verhamme P, Flameng W, Van de Werf F, Adriaenssens<br />

T. Comparison of thrombolysis versus surgery as a first line therapy<br />

for prosthetic heart valve thrombosis. Am J Cardiol 2011;107:275-<br />

279.<br />

54. Gupta D, Kothari SS, Bahl VK, Goswami KC, Talwar KK,<br />

Manchanda SC, Venugopal P. Thrombolytic therapy for prosthetic<br />

valve thrombosis: short- and long-term results. Am Heart J<br />

2000;140:906-916.<br />

55. Cáceres-Lóriga FM, Pérez-López H, Morlans-Hernández K,<br />

Facundo-Sánchez H, Santos-Gracia J, Valiente-Mustelier J,<br />

Rodiles-Aldana F, Marrero-Mirayaga MA, Betancourt BY, López-<br />

Saura P. Thrombolysis as first choice therapy in prosthetic heart<br />

valve thrombosis. A study of 68 patients. J Thromb Thrombolysis<br />

2006;21:185-190.<br />

56. Lengyel M, Vandor L. The role of thrombolysis in the management<br />

of left-sided prosthetic valve thrombosis: a study of 85 cases<br />

diagnosed by transesophageal echocardiography. J Heart Valve Dis<br />

2001;10:636-649.<br />

57. Ozkan M, Kaymaz C, Kirma C, Sönmez K, Ozdemir N, Balkanay<br />

M, Yakut C, Deligönül U. Intravenous thrombolytic treatment<br />

of mechanical prosthetic valve thrombosis: a study using<br />

serial transesophageal echocardiography. J Am Coll Cardiol<br />

2000;35:1881-1889.<br />

58. Shapira Y, Herz I, Vaturi M, Porter A, Adler Y, Birnbaum Y,<br />

Strasberg B, Sclarovsky S, Sagie A. Thrombolysis is an effective<br />

and safe therapy in stuck bileaflet mitral valves in the absence of<br />

high-risk thrombi. J Am Coll Cardiol 2000;35:1874-1880.<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

59. Cáceres-Lóriga FM, Pérez-López H, Santos-Gracia J, Morlans-<br />

Hernández K, Marrero-Mirayaga MA. Thrombolytic treatment as<br />

first option in recurrent tricuspid prosthetic valve thrombosis and<br />

Ebstein›s anomaly. J Pharm Pharm Sci 2005;8:332-334.<br />

60. Torrado González E, Ferriz Martín JA, Prieto Palomino MA,<br />

Rodríguez García JJ, Alvarez Bueno JM, Vera Almazán A, Garrido<br />

Alcalde MR, González de Vega N. Thrombolysis of thrombosed<br />

heart valve prostheses: presentation of 2 cases and review of the<br />

literature. Rev Esp Cardiol 1990;43:345-351.<br />

61. Shapira Y, Herz I, Birnbaum Y, Snir E, Vidne B, Sagie A. Repeated<br />

thrombolysis in multiple episodes of obstructive thrombosis in<br />

prosthetic heart valves: a report of three cases and review of the<br />

literature. J Heart Valve Dis 2000;9:146-149.<br />

62. Shapira Y, Vaturi M, Hasdai D, Battler A, Sagie A. The safety and<br />

efficacy of repeated courses of tissue-type plasminogen activator in<br />

patients with stuck mitral valves who did not fully respond to the<br />

initial thrombolytic course. J Thromb Haemost 2003;1:725-728.<br />

63. Ratnatunga CP, Edwards MB, Dore CJ, Taylor KM. Tricuspid<br />

valve replacement: UK Heart Valve Registry mid-term results<br />

comparing mechanical and biological prostheses. Ann Thorac Surg<br />

1998;66:1940-1947.<br />

64. Rizzoli G, Vendramin I, Nesseris G, Bottio T, Guglielmi C,<br />

Schiavon L. Biological or mechanical prostheses in tricuspid<br />

position A meta-analysis of intra-institutional results. Ann Thorac<br />

Surg 2004;77:1607-1614.<br />

65. Rizzoli G, De Perini L, Bottio T, Minutolo G, Thiene G, Casarotto<br />

D. Prosthetic replacement of the tricuspid valve: biological or<br />

mechanical Ann Thorac Surg 1998;66:S62-67.<br />

66. Kaplan M, Kut MS, Demirtas MM, Cimen S, Ozler A. Prosthetic<br />

replacement of tricuspid valve: bioprosthetic or mechanical. Ann<br />

Thorac Surg 2002;73:467-473.<br />

67. Chang BC, Lim SH, Yi G, Hong YS, Lee S, Yoo KJ, Kang MS,<br />

Cho BK. Long-term clinical results of tricuspid valve replacement.<br />

Ann Thorac Surg 2006;81:1317-1323.<br />

68. Filsoufi F, Anyanwu AC, Salzberg SP, Frankel T, Cohn LH, Adams<br />

DH. Long-term outcomes of tricuspid valve replacement in the<br />

current era. Ann Thorac Surg 2005;80:845-850.<br />

69. Dalrymple-Hay MJ, Leung Y, Ohri SK, Haw MP, Ross JK, Livesey<br />

SA, Monro JL. Tricuspid valve replacement: bioprostheses are<br />

preferable. J Heart Valve Dis 1999;8:644-648.<br />

70. Carrier M, Hébert Y, Pellerin M, Bouchard D, Perrault LP, Cartier<br />

R, Basmajian A, Pagé P, Poirier NC. Tricuspid valve replacement:<br />

an analysis of 25 years of experience at a single center. Ann Thorac<br />

Surg 2003;75:47-50.<br />

The Journal of Tehran University Heart Center155


The Journal of Tehran University Heart Center<br />

Original Article<br />

Increased Carotid Artery Intima-Media Thickness in<br />

Pregnant Women with Gestational Diabetes Mellitus<br />

Gholamreza Yousefzadeh, MD 1 , Hashem Hojat, MD 1 , Ahmad Enhesari, MD 1 ,<br />

Mostafa Shokoohi, MSc 2* , Nahid Eftekhari, MD 1 , Mehrdad Sheikhvatan, MD 3<br />

1<br />

Physiology Research Center, Kerman University of Medical Sciences, Kerman, Iran.<br />

2<br />

Research Center for Modeling in Health, Kerman University of Medical Sciences, Kerman, Iran.<br />

3<br />

Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.<br />

Received 04 February 2012; Accepted 13 May 2012<br />

Abstract<br />

Background: Pregnant women with previous gestational diabetes mellitus are at increased risk of progressive carotid<br />

artery disorders. The current study evaluated carotid intima-media thickness (IMT) in pregnant women with gestational<br />

diabetes at two time points of mid-term and full-term pregnancy to determine whether gestational diabetes mellitus causes<br />

increased IMT.<br />

Methods: This cross-sectional study carried out at Afzalipour Hospital (Kerman, Iran) between 2009 and 2010, recruited<br />

50 women who were at high risk of gestational diabetes during pregnancy and had an oral glucose challenge test (OGCT)<br />

as screening for gestational diabetes. B-mode ultrasound scans were performed at baseline and at two time points of midterm<br />

pregnancy (20 to 24 weeks) and full-term pregnancy (36 to 38 weeks) on all the participants. The mean IMT of common<br />

carotids and internal carotid arteries from two walls (near and far walls) at four different angles was assessed.<br />

Results: An overall comparison between the impaired OGCT test group and the control group revealed significant<br />

differences in carotid IMT in the mid-term (0.65 ± 0.07 vs. 0.59 ± 0.06 mm; p value = 0.002) and full-term (0.65 ± 0.05<br />

vs. 0.59 ± 0.04 mm; p value < 0.001) pregnancy; however, the trend of the changes in carotid IMT during mid to full-term<br />

pregnancy was insignificant in each group (p value > 0.05).<br />

Conclusion: Carotid IMT was significantly higher in the women with gestational diabetes than that in the normoglycemic<br />

group in different trimesters. This finding denotes that atherosclerosis might start years before the diagnosis of gestational<br />

diabetes in vulnerable women.<br />

J Teh Univ Heart Ctr 2012;7(4):156-159<br />

This paper should be cited as: Yousefzadeh G, Hojat H, Enhesari A, Shokoohi M, Eftekhari N, Sheikhvatan M. Increased Carotid<br />

Artery Intima-Media Thickness in Pregnant Women with Gestational Diabetes Mellitus. J TehUniv Heart Ctr 2012;7(4):156-159.<br />

Keywords: Pregnancy • Diabetes mellitus • Carotid arteries<br />

Introduction<br />

Screening for gestational diabetes mellitus is routinely<br />

programmed to prevent complications caused by elevated<br />

blood glucose levels in pregnancy, including macrosomia,<br />

Cesarean delivery, shoulder dystocia, neonatal metabolic<br />

problems, perinatal mortality, and pre-eclampsia. 1 Diabetes<br />

mellitus during pregnancy also appears to be associated<br />

*<br />

Corresponding Author: Mostafa Shokoohi, Research Center for Modeling in Health, Kerman University of Medical Sciences, Jihad Blvd, Shariati<br />

Street, Azadi Square, Kerman, Iran. 7619813159. Tel: +98 341 2263983. Fax: +98 341 2264079. E-mail: shokouhi.mostafa@gmail.com.<br />

156


Increased Carotid Artery Intima-Media Thickness in Pregnant Women with ...<br />

with an increased risk of cardiovascular diseases even in<br />

later life. 2 Moreover, this underlying co-morbidity is an<br />

independent risk factor for subsequent coronary artery<br />

disease. In this context, diabetes mellitus during pregnancy<br />

has been identified as a major etiology for atherosclerosis in<br />

large elastic arteries. 3<br />

One of the main indicators for assessing atherosclerotic<br />

lesions during this period is the increased intima-media<br />

thickness (IMT) of carotid arteries. 4 It has been well known<br />

that, as opposed to the blood flow through the other arteries of<br />

the maternal organs, the blood flow through the carotid artery<br />

is decreased during pregnancy, which has been attributed to<br />

pregnancy-mediated increased responsiveness of the carotid<br />

artery to vasoconstrictors and decreased responsiveness<br />

to vasodilators. 5 Also, the endothelial hypertrophy of the<br />

carotid artery can be a common finding, leading to changes<br />

in the carotid blood flow and its complications. 6, 7 Thus,<br />

increased carotid IMT can be deemed a validated endothelial<br />

dysfunction surrogate endpoint during pregnancy.<br />

Some studies have recently shown that pregnant women<br />

with previous gestational diabetes mellitus are at increased<br />

risk of carotid artery disorders. 8, 9 Be that as it may, the scarcity<br />

of research into this hypothesis means that it is still unknown<br />

whether or not gestational diabetes mellitus causes increased<br />

IMT. The current study was performed to evaluate carotid<br />

IMT in diabetic pregnant women with gestational diabetes<br />

and to ascertain if an impaired oral glucose challenge test<br />

(OGCT) correlates with the development of increased IMT.<br />

Methods<br />

This cohort study included 50 women ranging from<br />

18 to 35 years of age at high risk of diabetes during<br />

pregnancy. Gestational diabetes mellitus was screened<br />

with a one-hour 50 g oral glucose challenge test (OGCT).<br />

Abnormal results were, thereafter, confirmed with a threehour<br />

100 g oral glucose tolerance test (OGTT). Eligible<br />

women were nulliparous with a singleton pregnancy, had<br />

a normal blood pressure at the time of recruitment, and<br />

gave informed consent. Women with any of the following<br />

were excluded: family history of cardiovascular disorders;<br />

history of hypertension; anti-hypertensive and cholesterol<br />

medication use; hyperlipidemia; overt diabetes or fasting<br />

plasma glucose (FPG) > 125 mg/dl according to the<br />

American Diabetes Association (ADA) definition; 10 chronic<br />

renal or hepatic diseases; malignancies; recent hormonal<br />

medications; cigarette smoking; severe obesity (body<br />

mass index [BMI] > 35 kg/m 2 ); and history of infertility<br />

or polycystic ovarian disease. Those with the status of<br />

plaques/shadowing ( > 1.0 mm) at any carotid site were also<br />

excluded. The study protocol was approved by the Research<br />

and Ethics Committees at Kerman University of Medical<br />

Sciences.<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Baseline demographic variables were collected either<br />

from the women’s medical records or self-completed<br />

questionnaires at trial entry and comprised maternal age,<br />

height, weight, BMI, smoking status, and blood pressure<br />

at trial entry. Complete baseline data were available for<br />

all the women. All lipid and lipoprotein measurements<br />

were made at a central laboratory. Total cholesterol was<br />

measured enzymatically with standard methods and total<br />

triglyceride was measured via standard spectrophotometric<br />

techniques. After the precipitation of low-density lipoprotein<br />

(LDL) particle with phosphotungstic acid, high-density<br />

lipoprotein (HDL) cholesterol was measured enzymatically<br />

in the supernatant by a modification of the method for total<br />

cholesterol.<br />

B-mode ultrasound scans were performed at baseline<br />

and at two time points of mid-term pregnancy (20 to 24<br />

weeks) and full-term pregnancy (36 to 38 weeks) on all the<br />

participants. Carotid ultrasound scans were carried out by a<br />

single trained sonographer unaware of the study protocols<br />

and methodology. B-mode ultrasound images were equipped<br />

with a 7.5-MHz linear array transducer and captured with<br />

a GE log 200 ultrasound machine. The ultrasonic variable<br />

used in the statistical analysis was the mean of the IMT of<br />

common carotids and internal carotid arteries from two walls<br />

(near and far walls) at four different angles.<br />

The mean of carotid IMT was compared between the<br />

women with impaired OGCT and those who screened<br />

normal on OGCT at the two study time points. For the<br />

statistical analyses, the statistical software SPSS version<br />

19.0 for Windows (SPSS Inc., Chicago, IL) was used. The<br />

continuous variables, if normally distributed, were analyzed<br />

using the Student-test and presented as mean differences,<br />

while the Mann-Whitney test was employed for skewed<br />

data. A p value of 0.05 or less was considered to indicate<br />

statistical significance.<br />

Results<br />

The baseline characteristics of the pregnant women with<br />

impaired and normal OGCT tests are presented in Table 1.<br />

The two groups were similar in terms of pregnancy age,<br />

height, weight, BMI, and baseline laboratory parameters.<br />

There were no significant associations between carotid<br />

IMT and maternal indices, including the demographic<br />

variables and laboratory parameters.<br />

An overall comparison between the impaired OGCT test<br />

group and the control group via the Mann-Whitney U test<br />

revealed significant differences in carotid IMT in the midterm<br />

and full-term pregnancy (Table 2). The trend of the<br />

changes in carotid IMT during the mid-term to full-term<br />

pregnancy was, however, insignificant in each group.<br />

The Journal of Tehran University Heart Center157


The Journal of Tehran University Heart Center<br />

Gholamreza Yousefzadeh et al.<br />

Table 1. Demographic characteristics and clinical data of GDM versus non-GDM women *<br />

Characteristics<br />

GDM group<br />

(n=25)<br />

Non-GDM group<br />

(n=25)<br />

Age (y) 24.4±3.6 25.1±4.2 0.259<br />

Height (cm) 159.4±5.5 161.5±7.3 0.276<br />

Weight (kg) 72.8±11.5 68.7±10.9 0.225<br />

Body mass index (kg/m 2 ) 28.7±4.5 26.5±4.5 0.109<br />

Systolic BP (mm Hg) 110.0±9.4 107.6±8.0 0.369<br />

Diastolic BP (mm Hg) 69.0±10.4 65.2±8.5 0.192<br />

Serum HDL (mg/dL) 49.0±8.7 49.0±10.5 0.987<br />

Serum LDL (mg/dL) 140.7±37.4 137.7±36.0 0.780<br />

Fasting blood sugar (mg/dL) 85.3±13.7 83.2±19.8 0.680<br />

Serum triglyceride (mg/dL) 183.5±67.0 178.0±61.0 0.775<br />

Serum cholesterol (mg/dL) 219.0±54.7 223.8±43.8 0.746<br />

*<br />

Data are presented as mean±SD<br />

GDM, Gestational diabetes mellitus; BP, Blood pressure; HDL, High-density lipoprotein; LDL, Low-density lipoprotein<br />

P value<br />

Table 2. Differences in carotid IMT in GDM versus non-GDM women *<br />

CIMT<br />

Mid-term pregnancy<br />

(mm)<br />

Full-term<br />

pregnancy(mm)<br />

GDM group<br />

(n=25)<br />

Non-GDM group<br />

(n=25)<br />

P value **<br />

0.65±0.07 0.59±0.06 0.002<br />

0.65±0.05 0.59±0.04 < 0.001<br />

P value *** 0.989 0.992<br />

*<br />

Data are presented as mean±SD<br />

**<br />

Between groups comparison<br />

***<br />

Within groups comparison<br />

IMI, Intima-media thickness; GDM, Gestational diabetes mellitus; CIMT,<br />

Carotid intima-media thickness<br />

Discussion<br />

An increased carotid IMT can be observed not only in<br />

long-standing type II diabetic but also in newly detected type<br />

II diabetic patients. Especially in pregnant women, carotid<br />

IMT gradually increases from the first to the third trimester<br />

of normal pregnancy and regresses in the postpartum period.<br />

Nevertheless, the trend of the increase in pregnant women<br />

with gestational diabetes is still unclear. In our study, with the<br />

aid of carotid ultrasound, arterial examination was performed<br />

at mid-term and full-term pregnancy and both controls and<br />

the women with an impaired OGCT test were assessed<br />

and compared with regard to the changes in carotid IMT.<br />

We showed that although carotid IMT was more increased<br />

in those with impaired OGCT, the trend of this change was<br />

similar between the impaired and normal OGCT groups.<br />

Furthermore, we observed that carotid IMT was significantly<br />

higher in the diabetic group than in the normoglycemic<br />

group in different trimesters, but the trend of these gradual<br />

changes from the first to the third trimester was similar in<br />

both groups. Totally, effective glycemic control seems to be<br />

helpful for the prevention of increased carotid IMT.<br />

During the pregnancy period, peripheral vascular<br />

resistance is physiologically paralleled by improved<br />

macrovascular compliance. 11, 12 Some researchers have<br />

shown that 2-4 years after previous gestational diabetes<br />

mellitus, a significantly higher arterial stiffness can be<br />

found compared with reference women without a history of<br />

diabetes. 13 Pregnancies complicated by diabetes not only are<br />

associated with increased carotid IMT but also can lead to<br />

increased maternal arterial stiffness. 9<br />

This study demonstrates increased IMT of the carotid artery<br />

in a group of women who had a pregnancy complicated by<br />

impaired OGCT. This finding is in line with the literature<br />

confirming that women with a history of gestational diabetes<br />

have a higher risk of developing increased carotid IMT. This<br />

change can potentially lead to cardiovascular diseases in<br />

later life. There are some explanations for this phenomenon.<br />

One of the first steps in the development of atherosclerosis is<br />

endothelial activation, followed by endothelial dysfunction.<br />

Thus, in diabetes mellitus, vascular endothelial activation<br />

or dysfunction is considered to play a key role in the<br />

development of many of the clinical manifestations related<br />

to carotid artery disease, several years after delivery.<br />

Association between parity and carotid artery disease<br />

can be affected by some underlying factors. Some<br />

epidemiological studies have found positive associations<br />

between parity and risk of carotid artery plaques in elderly<br />

women and, therefore, age of pregnancy can be a trigger<br />

for this phenomenon. 14 Also, multi-gravidity and previous<br />

history of coronary diseases have been also identified to be<br />

related to the appearance of carotid artery plaques. 15 Folate<br />

deficiency during pregnancy is another probable triggering<br />

factor for progressing carotid IMT. Folate deficiency is<br />

linked with hyperhomocystinemia, 16 which is associated with<br />

accelerated progression of atherosclerosis 17 and, thereby,<br />

158


Increased Carotid Artery Intima-Media Thickness in Pregnant Women with ...<br />

carotid artery IMT. Pregnancy is potentially a cause of folate<br />

deficiency. 18 Consequently, it is possible that multiparous<br />

women are at risk of prolonged folate deficiency; this may<br />

explain their susceptibility to the development of carotid<br />

artery IMT.<br />

In the present study, the main limitation was blood glucose<br />

controlled by insulin; we could not hold treatment because<br />

of ethical issues. Also, we did not assess the confounding<br />

effects of the triggering variables such as age of pregnancy,<br />

multi-parity, or medication during this period on increasing<br />

carotid IMT; that should be considered in further studies.<br />

Another weakness of this study would be the relatively small<br />

sample size. The study was designed to enroll 50women.<br />

Despite this, we feel that it provides valuable information.<br />

Prospective follow-up studies will also be needed and have<br />

meanwhile been started to determine whether diabetes during<br />

pregnancy itself is responsible for the increase in IMT.<br />

Conclusion<br />

In conclusion, an impaired OGCT test is proven to be<br />

an independent risk factor for increased carotid IMT and<br />

subsequent coronary artery disease. Even with this small<br />

study, we were able to find an increased IMT after diabetes<br />

appearance, which might be used as an indicator of a potential<br />

increased vascular risk. Furthermore, IMT measurements<br />

in diabetic pregnant women could offer an opportunity to<br />

identify a high-risk group of women who might benefit from<br />

early screening and preventive measures. These measures<br />

could include lifestyle interventions such as improving diet<br />

and physical activity as well as increased surveillance of<br />

blood pressure, serum lipids, and particularly blood glucose.<br />

Acknowledgment<br />

This study was a resident’s thesis and was financially<br />

supported by Kerman University of Medical Sciences.<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

artery intima-media thickness in cardiovascular risk assessment by<br />

Framingham risk-score in type 2 diabetes: a retrospective cohort<br />

study. J Diabetes 2009;1:188-193.<br />

5. Akinci B, Demir T, Celtik A, Baris M, Yener S, Ozcan MA, Yuksel<br />

F, Secil M, Yesil S. Serum osteoprotegerin is associated with<br />

carotid intima media thickness in women with previous gestational<br />

diabetes. Diabetes Res Clin Pract 2008;82:172-178.<br />

6. Blaauw J, van Pampus MG, Van Doormaal JJ, Fokkema MR, Fidler<br />

V, Smit AJ, Aarnoudse JG. Increased intima-media thickness after<br />

early-onset preeclampsia. Obstet Gynecol 2006;107:1345-1351.<br />

7. Jovanović S, Jovanović A. Pregnancy is associated with hypotrophy<br />

of carotid artery endothelial and smooth muscle cells. Hum Reprod<br />

1998;13:1074-1078.<br />

8. Tarim E, Yigit F, Kilicdag E, Bagis T, Demircan S, Simsek<br />

E, Haydardedeoglu B, Yanik F. Early onset of subclinical<br />

atherosclerosis in women with gestational diabetes mellitus.<br />

Ultrasound Obstet Gynecol 2006;27:177-182.<br />

9. Savvidou MD, Anderson JM, Kaihura C, Nicolaides KH. Maternal<br />

arterial stiffness in pregnancies complicated by gestational and<br />

type 2 diabetes mellitus. Am J Obstet Gynecol 2010;203:274.e1-7.<br />

10. American Diabetes Association. Standards of medical care in<br />

diabetes-2011. Diabetes Care2011;34:11-61.<br />

11. Visontai Z, Lenard Z, Studinger P, Rigo J, Jr, Kollai M. Impaired<br />

baroreflex function during pregnancy is associated with stiffening<br />

of the carotid artery. Ultrasound Obstet Gynecol 2002;20:364-369.<br />

12. Mersich B, Rigó J, Jr, Besenyei C, Lénárd Z, Studinger P, Kollai M.<br />

Opposite changes in carotid versus aortic stiffness during healthy<br />

human pregnancy. Clin Sci 2005;109:103-107.<br />

13. Hu J, Norman M, Wallensteen M, Gennser G. Increased large<br />

arterial stiffness and impaired acetylcholine induced skin<br />

vasodilatation in women with previous gestational diabetes<br />

mellitus. Br J Obstet Gynaecol 1998;105:1279-1287.<br />

14. Humphries KH, Westendorp IC, Bots ML, Spinelli JJ, Carere<br />

RG, Hofman A. Parity and carotid artery atherosclerosis inelderly<br />

women: the Rotterdam Study. Stroke2001;32:2259-2264.<br />

15. Ness RB, Harris T, Cobb J, Flegal KM, Kelsey JL, Balanger A.<br />

Number of pregnancies and the subsequent risk of cardiovascular<br />

disease. N Engl J Med 1993;328:1528-1533.<br />

16. Boushey CJ, Beresford SA, Omenn GS, Motulsky AG. A<br />

quantitative assessment of plasma homocysteine as a risk factor for<br />

vascular disease. Probable benefits of increasing folic acid intakes.<br />

JAMA 1995;274:1049-1057.<br />

17. Selhub J, Jacques PF, Bostom AG, D’Agostino RB, Wilson PW,<br />

Belanger AJ, O’Leary DH, Wolf PA, Schaefer EJ, Rosenberg IH.<br />

Association between plasma homocysteine concentrations and<br />

extracranial carotid-artery stenosis. N Engl J Med1995;332:286-<br />

291.<br />

18. Mc Partlin J, Halligan A, Scott JM, Darling M, Weir DG. Accelerated<br />

folate breakdown in pregnancy. Lancet 1993;341:148-149.<br />

References<br />

1. Bo S, Valpreda S, Menato G, Bardelli C, Botto C, Gambino R,<br />

Rabbia C, Durazzo M, Cassader M, Massobrio M, Pagano G.<br />

Should we consider gestational diabetes a vascular risk factor<br />

Atherosclerosis 2007;194:72-79.<br />

2. Ben-Haroush A, Yogev Y, Hod M. Epidemiology of gestational<br />

diabetes mellitus and its association with type 2 diabetes. Diabet<br />

Med 2004;21:103-113.<br />

3. Vastagh I, Horváth T, Garamvölgyi Z, Rosta K, Folyovich A, Rigó<br />

J, Kollai M, Bereczki D, Somogyi A. Preserved structural and<br />

functional characteristics of common carotid artery in properly<br />

treated normoglycemic women with gestational diabetes mellitus.<br />

Acta Physiol Hung 2011;98:294-304.<br />

4. Ataoglu HE, Saler T, Uzunhasan I, Yenigun M, Yigit Z, Temiz LU,<br />

Saglam Z, Cetin F, Kumbasar B, Sar F. Additional value of carotid<br />

The Journal of Tehran University Heart Center159


The Journal of Tehran University Heart Center<br />

Original Article<br />

Right Ventricular Myocardial Tissue Velocities,<br />

Myocardial Performance Index, and Tricuspid Annular<br />

Plane Systolic Excursion in Totally Corrected Tetralogy<br />

of Fallot Patients<br />

Asadolah Tanasan, MD 1 , Keyhan Sayadpour Zanjani, MD 2* , Armen<br />

Kocharian, MD 2 , Abdolrazagh Kiani, MD 2 , Mohammad Ali Navabi, MD 2<br />

1<br />

Besat Hospital, Hamadan University of Medical Sciences, Hamadan, Iran.<br />

2<br />

Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran.<br />

Received 03 November 2011; Accepted 16 May 2012<br />

Abstract<br />

Background: Longer survival after the total repair of the Tetralogy of Fallot increases the importance of late complications<br />

such as right ventricular dysfunction. This is a prospective study of the right ventricular function in totally corrected Tetralogy<br />

of Fallot patients versus healthy children.<br />

Methods: Thirty-two healthy children were prospectively compared with 30 totally corrected Tetralogy of Fallot patients.<br />

Right ventricular myocardial tissue velocities, right ventricular myocardial performance index, and tricuspid annular plane<br />

systolic excursion were investigated as well as the presence and severity of pulmonary regurgitation.<br />

Results: The two groups were age-and sex-matched. Mean systolic peak velocity (Sa) and tricuspid annular plane systolic<br />

excursion were significantly decreased, while myocardial performance index and early to late diastolic velocity (Ea/Aa)<br />

were significantly increased in the Tetralogy of Fallot patients. Early diastolic velocity (Ea) showed no significant difference<br />

between the two groups. Sa correlated significantly with tricuspid annular plane systolic excursion in both the normal children<br />

and totally corrected Tetralogy of Fallot patients. Myocardial performance index was significantly higher in the patients<br />

with moderate to severe pulmonary regurgitation than in those with mild regurgitation. However, there was no significant<br />

correlation between this index and right ventricular myocardial tissue velocities.<br />

Conclusion: In this study, systolic right ventricular function indices (Sa and tricuspid annular plane systolic excursion)<br />

were impaired in the totally corrected Tetralogy of Fallot patients. Myocardial performance index was affected by the severity<br />

of pulmonary regurgitation.<br />

J Teh Univ Heart Ctr 2012;7(4):160-163<br />

This paper should be cited as: Tanasan A, Sayadpour Zanjani K, Kocharian A, Kiani A, Navabi MA. Right Ventricular Myocardial<br />

Tissue Velocities, Myocardial Performance Index, and Tricuspid Annular Plane Systolic Excursion in Totally Corrected Tetralogy of Fallot<br />

Patients. J Teh Univ Heart Ctr 2012;7(4):160-163.<br />

Keywords: Tetralogy of Fallot • Tricuspid valve • Elasticity imaging technique • Child • Heart ventricles<br />

*<br />

Corresponding Author: Keyhan Sayadpour Zanjani, Assistant Professor of Pediatric Cardiology, Tehran University of Medical Sciences, Children’s<br />

Medical Center, No.62, Dr Gharib Street, Tehran, Iran. 1419733151. Tel: +98 21 66911029. Fax: +98 21 66930024. E-mail: sayadpour@tums.ac.ir.<br />

160


Right Ventricular Myocardial Tissue Velocities, Myocardial Performance Index ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Introduction<br />

Although excellent outcomes have been achieved long<br />

after the total correction of the Tetralogy of Fallot (T<strong>OF</strong>) in<br />

the developed countries, right ventricular (RV) dysfunction<br />

may still cause mortality and morbidity. 1, 2 Pulmonary<br />

regurgitation (PR) has a detrimental effect on the RV<br />

diastolic function. 3 Late systolic RV dysfunction has been<br />

reported after the correction of the T<strong>OF</strong> in the developed<br />

countries. 4 The long-term survival after the T<strong>OF</strong> repair in the<br />

developing countries is unknown; 5, 6 we, therefore, sought to<br />

focus on the earlier RV function in these patients. We used<br />

the right ventricular myocardial performance index (RVMPI<br />

or Tei index), tricuspid annular plane systolic excursion<br />

(TAPSE), and myocardial tissue velocities for the evaluation<br />

of the RV systolic and diastolic functions in totally corrected<br />

T<strong>OF</strong> patients. 7<br />

Methods<br />

Thirty consecutive totally corrected T<strong>OF</strong> patients, who<br />

referred to our Outpatient Clinic between July 2008 and<br />

July 2009, were enrolled in the study and written informed<br />

consent was obtained from all of them. The study population<br />

underwent surgery using valved trans-annular patches, nonvalved<br />

trans-annular patches, or sub-annular patches at<br />

a mean age of 3.6 ± 3.2 years. Thirty-two normal healthy<br />

children with innocent murmur were selected as the control<br />

group.<br />

The inclusion criteria for this study included classic T<strong>OF</strong><br />

with pulmonary stenosis. The exclusion criteria were other<br />

variants of the T<strong>OF</strong> (pulmonary atresia with ventricular septal<br />

defect [VSD], absent pulmonary valve, and double-outlet<br />

RV) and the presence of a significant residual abnormality<br />

(VSD, valvular or peripheral pulmonary stenosis, or tricuspid<br />

regurgitation) other than PR.<br />

Echocardiography was performed using a single<br />

echocardiographic scanner (MicroMaxx Ultrasound System,<br />

Sonosite Inc., USA). RVMPI was measured using continuous<br />

pulse wave Doppler of the pulmonary and tricuspid valves.<br />

The formula a-b/b was used, where a was the time interval<br />

between cessation and onset of tricuspid inflow and b was<br />

pulmonary ejection time. 7<br />

The systolic displacement of the lateral portion of the<br />

tricuspid annular plan (TAPSE) was measured on the<br />

M-mode tracing under a two-dimensional echocardiogram<br />

in the four-chamber view. 8<br />

The averages of the RV systolic (Sa), early diastolic (Ea),<br />

and late diastolic velocities (Aa) in three cardiac cycles were<br />

measured using tissue Doppler echocardiography. 4 Threemillimeter<br />

Doppler sample volume was applied parallel<br />

to the RV free wall at the tricuspid valvular level to avoid<br />

sampling the right-heart cavities due to normal cardiac<br />

movement.<br />

Severity of PR was assessed by color and continues<br />

Doppler studies in the parasternal short-axis view. Duration<br />

of the PR flow to total diastolic time (PR index and PRi)<br />

was measured. PRi ≥ 70% was defined as mild PR, while<br />

9, 10<br />

the lower values were defined as moderate to severe PR.<br />

The severity of PR was also assessed on the color Doppler<br />

imaging. The presence of persistent retrograde color flow<br />

from the distal main pulmonary artery or branches that<br />

extend into the RV was defined as moderate to severe PR. 3<br />

The study was approved by the local Ethics Committee and<br />

it was conducted in accordance with good clinical practice<br />

and the Declaration of Helsinki.<br />

For the statistical analyses, the statistical software<br />

SPSS version 13.0 for Windows (SPSS Inc., Chicago, IL)<br />

was used. An independent samples t-test was performed<br />

to compare the RV function indices between the normal<br />

children and T<strong>OF</strong> patients. A bivariate correlation was<br />

performed using the Pearson correlation coefficient<br />

between the RV function indices. Statistical significance<br />

was inferred at a p value < 0.05.<br />

Results<br />

The 32 normal healthy children (17 males and 15 females<br />

at a mean age of 5.6 ± 3.8 years) and the 30 totally corrected<br />

T<strong>OF</strong> patients (17 males and 13 females at a mean age of<br />

6.4 ± 4.1 years) were age- and sex-matched. PR severity<br />

was evaluated in all of the totally corrected T<strong>OF</strong> patients,<br />

of whom 15 had valved trans-annular, 8 non-valved transannular,<br />

and 7 sub-annular patches.<br />

There were statistically significant differences in RVMPI,<br />

TAPSE, and myocardial tissue velocities (Sa, Aa, and Ea/<br />

Aa) between the healthy children and totally corrected T<strong>OF</strong><br />

patients. There was no significant difference in Ea (Table 1).<br />

Table 1. Indices of the RV function in the totally corrected T<strong>OF</strong> patients and<br />

normal healthy children *<br />

Variable T<strong>OF</strong> group Normal group P value<br />

RVMPI 0.3±0.1 0.2±0.1 0.001<br />

TAPSE (mm) 13.6±2.9 16.3±3.3 0.001<br />

Sa (cm/s) 8.6±8.9 11.4±2.2 < 0.001<br />

Ea (cm/s) 10.9±3.3 11.9±2.1 > 0.05<br />

Aa (cm/s) 5.9±1.8 8.2±2.1 < 0.001<br />

Ea/Aa 2.0±0.8 1.5±0.4 0.015<br />

*<br />

Data are presented as mean±SD<br />

RV, Right ventricle; T<strong>OF</strong>, Tetralogy of Fallot; RVMPI, Right ventricular<br />

myocardial performance index; TAPSE, Tricuspid annular plain systolic<br />

excursion; Sa, RV systolic velocity; Ea, Early diastolic velocity; Aa, Late<br />

diastolic velocity<br />

The Pearson analysis showed a strong correlation between<br />

The Journal of Tehran University Heart Center161


The Journal of Tehran University Heart Center<br />

TAPSE and Sa both in the control (r = 0.755, p value < 0.001)<br />

and patient (r = 0.662, p value = 0.002) groups. There was<br />

a significant inverse correlation between Sa and RVMPI (p<br />

value = 0.001) in the control group but not in the patient<br />

group (p value = 0.054). There was no significant correlation<br />

between RVMPI and Ea/Aa in either group.<br />

PRi was measured in all the totally corrected T<strong>OF</strong> patients,<br />

of whom 13 had a value ≥ 70% (mild PR) and 17 had a value<br />

< 70% (moderate to severe PR). PRi < 50% was found in 2<br />

patients. There was a significant correlation between RVMPI<br />

and PRi (p value < 0.001) in the patient group. The RVMPI<br />

in patients with PRi < 70% was 0.42 ± 0.13, which differed<br />

significantly from the RVMPI (0.24 ± 0.1) in the patients<br />

with PRi ≥ 70% (p value < 0.05). In the 2 patients with PRi <<br />

50%, RVMPI was 0.62 and 0.47, respectively. There was no<br />

significant difference in the Ea/Aa ratio between the patients<br />

who had PRi ≥ 70% and those who had PRi < 70% (p value<br />

= 0.38). The Ea/Aa ratio was insignificantly higher in PRi<br />

< 70% (2.1 ± 9.4 vs. 1.85 ± 7.4; p value = 0.405). In the 2<br />

patients who had PRi < 50%, the Ea/Aa ratio was 4.57/1 and<br />

2.92/1, respectively (restrictive pattern).<br />

In the color flow Doppler assessment of PR, 11 patients<br />

had mild PR and 19 had moderate to severe PR. There was a<br />

significant correlation between PR severity in the retrograde<br />

color flow mapping and RVMPI (p value = 0.023), but there<br />

was no correlation between PR severity and Ea/Aa ratio (p<br />

value = 0.38).<br />

Discussion<br />

Advances in cardiac surgery and postoperative<br />

management in the developed countries have contributed<br />

to the excellent long-term outcome of the totally corrected<br />

T<strong>OF</strong> patients; 1 nevertheless, the long-term outcome of<br />

the corrected T<strong>OF</strong> surgery in developing countries is<br />

unknown. 5, 6 In the developed countries, PR and RV<br />

diastolic dysfunction is the most important outcome factor<br />

following the T<strong>OF</strong> repair. 2, 3 There are reports of the usage<br />

of pulmonary valve sparing surgery to decrease the severity<br />

of PR in the T<strong>OF</strong> patients. 11, 12 Intra- and postoperative<br />

management problems, delay in corrective surgery, and<br />

palliative surgery (aorto-pulmonary shunts) are probably<br />

additional contributory factors to the poor long-term<br />

outcome in developing countries. 5, 6 At follow-ups of the<br />

repaired T<strong>OF</strong> patients, simple and fast evaluation of the<br />

systolic and diastolic RV functions is of great importance.<br />

In our study, the RV systolic and diastolic function indices<br />

were clearly impaired in the T<strong>OF</strong> patients in comparison with<br />

the normal children at mid-term follow-ups (Table 1). A few<br />

studies have focused on the RV function at mid-term followup<br />

after the T<strong>OF</strong> repair. Pilla et al. evaluated the presence<br />

of mid-term RV dysfunction and health-related quality of<br />

life after the T<strong>OF</strong> repair in Brazil. 13 In their investigation,<br />

Asadolah Tanasan et al.<br />

35 patients at a median age of 6.1 years and 4.9 years of<br />

follow-up after surgery were compared with 36 sex- and<br />

age-matched healthy controls and RVMPI (0.34 vs. 0.2)<br />

demonstrated RV dysfunction in the patients compared with<br />

the controls. In adult patients late after the repair of the T<strong>OF</strong>,<br />

D’Andrea et al. detected a correlation between myocardial<br />

performance assessed at rest via tissue Doppler (TD) and<br />

cardiac performance during physical effort in adult patients<br />

(21.4 ± 3.8 years). 4 In their study, TD analysis showed lower<br />

Sa, Ea, Ea/Aa ratios in the corrected T<strong>OF</strong> patients compared<br />

to the normal group. In contrast to our study, the corrected<br />

T<strong>OF</strong> patients of their study had no significant PR, the mean<br />

age of the T<strong>OF</strong> patients was higher than that in our study<br />

(21.4 ± 3.8 vs. 6.3 ± 3.3 years), and the mean age of the<br />

corrected T<strong>OF</strong> patients was lower than that in our study (1.4<br />

± 0.5 vs. 3.6 ± 3.2 years). 4 In the Puranik et al. study, Sa<br />

and Ea were significantly lower in asymptomatic corrected<br />

adult T<strong>OF</strong> patients compared with their normal group. 14<br />

Similar to our study, there was significant PR but the age at<br />

corrective surgery was higher (6.6 vs. 3.6 years). Although<br />

the increased Ea/Aa ratio in our study, in contrast to the<br />

D’Andrea’s, may suggest the role of combined prolonged<br />

cyanosis and post-correction PR, a comparison with the<br />

Puranik study emphasizes the importance of other factors in<br />

4, 14<br />

the restrictive diastolic RV dysfunction in our patients.<br />

On the other hand, while our study demonstrates combined<br />

systolic dysfunction (decrease in both Sa and TAPSE) and<br />

restrictive RV dysfunction (increase in Ea/Aa ratio), the two<br />

other studies 4, 14 showed mild systolic dysfunction (decrease<br />

in Sa) associated with a significant decrease in Ea/Aa in adult<br />

corrected T<strong>OF</strong> patients. Frigola et al. reported that PR was an<br />

important factor of RV contractile dysfunction after the T<strong>OF</strong><br />

repair. 15 These observations may suggest that the systolic and<br />

restrictive diastolic RV dysfunctions in our study probably<br />

originated through others mechanisms and they were present<br />

since the early postoperative period. In a study by Cullen<br />

et al, 16 patients with early restrictive physiology after the<br />

T<strong>OF</strong> repair had a clinical picture of low cardiac output and<br />

slow postoperative recovery but a study by Gatzoulis et al.<br />

suggested that late restrictive physiology predicted a superior<br />

exercise performance. 17 Further studies are clearly warranted<br />

about how the RV systolic and diastolic functions correlate<br />

with intra- and early postoperative management and PR.<br />

In addition to significant PR and prolonged preoperative<br />

cyanosis, intra- or early postoperative management may<br />

have been affected by systolic and restrictive diastolic RV<br />

functions in our study. 6 Although RVMPI was significantly<br />

correlated with PR severity (retrograde color flow and PRi),<br />

there was no significant correlation between RVMPI and<br />

Ea/Aa ratio. In our 2 patients with severe PR (PRi < 50%),<br />

the Ea/Aa ratio was significantly elevated. As RVMPI was<br />

not correlated with systolic and diastolic function indices<br />

(Sa, TAPSE, Ea, and Ea/Aa) but was allied to PR severity,<br />

RVMPI may have been affected more by PR severity than<br />

162


Right Ventricular Myocardial Tissue Velocities, Myocardial Performance Index ...<br />

by the RV function in the totally corrected T<strong>OF</strong> patients with<br />

restrictive physiology. In accordance with our study, Abd<br />

El Rahman et al. suggested that MPI was affected by the<br />

severity of PR in the presence of RV diastolic dysfunction. 18<br />

Yasuoka et al. showed that MPI obtained by pulsed wave<br />

Doppler was not different in patients with the T<strong>OF</strong> repair<br />

compared with normal children but RVMPI measured by<br />

tissue Doppler was significantly higher in patients with the<br />

T<strong>OF</strong> than in normal children. 19<br />

The current study had some limitations. The sample<br />

size was inadequate to compare the totally corrected T<strong>OF</strong><br />

subgroups. Furthermore, half of the corrected T<strong>OF</strong> patients<br />

underwent valved trans-annular patch repair, which is a new<br />

technique for right ventricular outflow tract reconstruction.<br />

Conclusion<br />

RVMPI was significantly correlated with PR severity<br />

without the presence of a significant correlation between<br />

RVMPI (obtained by pulsed wave Doppler) and the RV<br />

function indices obtained by tissue Doppler imaging<br />

(EA, Aa, Ea/Aa, and Sa). We suggest that these indices,<br />

RVMPI by tissue Doppler and PRi, be measured basically<br />

at postoperative and follow-up evaluations. In addition, as<br />

TAPSE was significantly decreased in the totally corrected<br />

T<strong>OF</strong> patients and there was a significant correlation between<br />

TAPSE and Sa, we suggest that TAPSE be also measured as<br />

the global RV systolic function index.<br />

Acknowledgment<br />

This study was approved and supported by Tehran<br />

University of Medical Sciences.<br />

References<br />

1. Bacha EA, Scheule AM, Zurakowski D, Erickson LC, Hung J, Lang<br />

P, Mayer JE, Jr, del Nido PJ, Jonas RA. Long-term results after<br />

early primary repair of tetralogy of Fallot. J Thorac Cardiovasc<br />

Surg 2001;122:154-161.<br />

2. Knauth AL, Gauvreau K, Powell AJ, Landzberg MJ, Walsh<br />

EP, Lock JE, del Nido PJ, Geva T. Ventricular size and function<br />

assessed by cardiac MRI predict major adverse clinical outcomes<br />

late after tetralogy of Fallot repair. Heart 2008;94:211-216.<br />

3. Bouzas B, Kilner PJ, Gatzoulis MA. Pulmonary regurgitation: not<br />

a benign lesion. Eur Heart J 2005;26:433-439.<br />

4. D‘Andrea A, Caso P, Sarubbi B, Russo MG, Ascione L, Scherillo M,<br />

Cobrufo M, Calabrò R. Right ventricular myocardial dysfunction<br />

in adult patients late after repair of tetralogy of fallot. Int J Cardiol<br />

2004;94:213-220.<br />

5. Ho KW, Tan RS, Wong KY, Tan TH, Shankar S, Tan JL. Late<br />

complications following tetralogy of Fallot repair: the need for<br />

long-term follow-up. Ann Acad Med Singapore 2007;36:947-953.<br />

6. Rao SG. Pediatric cardiac surgery in developing countries. Pediatr<br />

Cardiol 2007;28:144-148.<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

7. Haddad F, Hunt SA, Rosenthal DN, Murphy DJ. Right ventricular<br />

function in cardiovascular disease. Part I: anatomy, physiology,<br />

aging, and functional assessment of the right ventricle. Circulation<br />

2008;117:1436-1448.<br />

8. Miller D, Farah MG, Liner A, Fox K, Schluchter M, Hoit BD. The<br />

relation between quantitative right ventricular ejection fraction and<br />

indices of tricuspid annular motion and myocardial performance. J<br />

Am Soc Echocardiogr 2004;17:443-447.<br />

9. Festa P, Ait-Ali L, Minichilli F, Kristo I, Deiana M, Picano E.<br />

A new simple method to estimate pulmonary regurgitation by<br />

echocardiography in operated fallot: comparison with magnetic<br />

resonance imaging and performance test evaluation. J Am Soc<br />

Echocardiogr 2010;23:496-503.<br />

10. Li W, Davlouros PA, Kilner PJ, Pennell DJ, Gibson D, Henein<br />

MY, Gatzoulis MA. Doppler-echocardiographic assessment of<br />

pulmonary regurgitation in adults with repaired tetralogy of Fallot:<br />

comparison with cardiovascular magnetic resonance imaging. Am<br />

Heart J 2004;147:165-172.<br />

11. Boni L, García E, Galletti L, Pérez A, Herrera D, Ramos V,<br />

Marianeschi SM, Comas JV. Current strategies in tetralogy of<br />

Fallot repair: pulmonary valve sparing and evolution of right<br />

ventricle/left ventricle pressures ratio. Eur J Cardiothorac Surg<br />

2009;35:885-889.<br />

12. Singh S, Pratap H, Agarwal S, Singh A, Satsangi DK. Pulmonary<br />

valve preservation in tetralogy of Fallot with a mildly hypoplastic<br />

annulus-should we do it Indian J Thorac Cardiovasc Surg<br />

2011;27:76-82.<br />

13. Pilla CB, Pereira CA, Fin AV, Aquino FV, Botta A, DalleMulle L,<br />

Ricachinevsky CP, Nogueira AJ, Lucchese FA, Rohde LE. Healthrelated<br />

quality of life and right ventricular function in the midterm<br />

follow-up assessment after tetralogy of Fallot repair. Pediatr<br />

Cardiol 2008;29:409-415.<br />

14. Puranik R, Greaves K, Hawker RE, Pressley LA, Robinson PJ,<br />

Celermajer DS. Abnormal right ventricular tissue velocities after<br />

repair of congenital heart disease-implications for late outcomes.<br />

Heart Lung Circ 2007;16:295-299.<br />

15. Frigiola A, Redington AN, Cullen S, Vogel M. Pulmonary<br />

regurgitation is an important determinant of right ventricular<br />

contractile dysfunction in patients with surgically repaired<br />

tetralogy of Fallot. Circulation 2004;110:II153-157.<br />

16. Cullen S, Shore D, Redington A. Characterization of right<br />

ventricular diastolic performance after complete repair of tetralogy<br />

of Fallot. Restrictive physiology predicts slow postoperative<br />

recovery. Circulation 1995;91:1782-1789.<br />

17. Gatzoulis MA, Clark AL, Cullen S, Newman CG, Redington AN.<br />

Right ventricular diastolic function 15 to 35 years after repair<br />

of tetralogy of Fallot. Restrictive physiology predicts superior<br />

exercise performance. Circulation 1995;91:1775-1781.<br />

18. Abd El Rahman MY, Abdul-Khaliq H, Vogel M, Alexi-Meskischvili<br />

V, Gutberlet M, Hetzer R, Lange PE. Value of the new Dopplerderived<br />

myocardial performance index for the evaluation of right<br />

and left ventricular function following repair of tetralogy of Fallot.<br />

Pediatr Cardiol 2002;23:502-507.<br />

19. Yasuoka K, Harada K, Toyono M, Tamura M, Yamamoto F. Tei<br />

index determined by tissue Doppler imaging in patients with<br />

pulmonary regurgitation after repair of tetralogy of Fallot. Pediatr<br />

Cardiol 2004;25:131-136.<br />

The Journal of Tehran University Heart Center163


The Journal of Tehran University Heart Center<br />

Original Article<br />

Anatomy of Atrioventricular Node Artery and Pattern of<br />

Dominancy in Normal Coronary Subjects: A Comparison<br />

between Individuals with and without Isolated Right<br />

Bundle Branch Block<br />

Ali Kazemisaeid, MD, Marziyeh Pakbaz, MD * , Ahmad Yaminisharif, MD,<br />

Gholamreza Davoodi, MD, Masoumeh Lotfi Tokaldany, MD, Elham Hakki<br />

Kazazi, MD<br />

Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.<br />

Received 24 December 2011; Accepted 20 May 2012<br />

Abstract<br />

Background: Isolated right bundle branch block (RBBB) is a common finding in the general population. The atrioventricular<br />

node (AVN) artery contributes to the blood supply of the right bundle branch. Our hypothesis was that the anatomy of the<br />

AVN artery and the pattern of dominancy differ between subjects with and without RBBB.<br />

Methods: We retrospectively studied the coronary angiography of 92 patients with RBBB and 184 age- and gendermatched<br />

controls without RBBB. All the subjects had angiographically proven normal coronary arteries. The dominant<br />

circulation and precise origin of the AVN artery were determined in each subject. Obtained data were compared between the<br />

two study groups.<br />

Results: There was no significant difference between the two groups in terms of dominancy (p value = 0.200). Origination<br />

of the AVN artery from the right circulatory system was more common in both groups, but this pattern was more prevalent<br />

in the cases than in the controls (p value = 0.021). There was a great variation of the AVN artery origin. In the total study<br />

population, the AVN artery was more commonly separated from a non crux origin than from the crux area. The prevalence of<br />

the non-crux origination of the AVN artery was significantly higher in the cases than in the controls (p value < 0.001). While<br />

the origination of the AVN artery from the right circulatory system was more common in both groups, the prevalence of the<br />

right origin of the AVN artery was significantly higher in the cases than in the controls. We observed that the AVN artery most<br />

commonly originated from the dominant artery but not necessarily from the crux.<br />

Conclusion: The anatomy of the AVN artery but not the pattern of dominancy is somewhat different in subjects with RBBB<br />

compared with normal individuals.<br />

J Teh Univ Heart Ctr 2012;7(4):164-169<br />

This paper should be cited as: Kazemisaeid A, Pakbaz M, Yaminisharif A, Davoodi G, Lotfi Tokaldany M, Hakki Kazazi E. Anatomy<br />

of Atrioventricular Node Artery and Pattern of Dominancy in Normal Coronary Subjects; A Comparison Between Individuals With and<br />

Without Isolated Right Bundle Branch Block. J TehUniv Heart Ctr 2012;7(4):164-169.<br />

Keywords: Anatomy • Coronary vessels • Bundle-branch block<br />

*<br />

Corresponding Author: Marziyeh Pakbaz, Department of Cardiology, Tehran University of Medical Sciences, Tehran Heart Center, North Kargar<br />

Street, Tehran, Iran.1411713138. Tel: +98 21 88029256. Fax: +98 21 88029256. E-Mail: marzi.pakbaz@gmail.com.<br />

164


Anatomy of Atrioventricular Node Artery and Pattern of Dominancy in Normal ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Introduction<br />

The atrioventricular node (AVN) is an area of specialized<br />

tissue between the atria and the ventricles of the heart and<br />

specifically acts as an electrical relay station for electrical<br />

impulses, passing from the atria to the ventricles. The arterial<br />

blood supply of the AVN, which is quite variable, was the<br />

topic of anatomic, histologic, and radiologic research for<br />

many years. The origin of the blood supply to the conduction<br />

system is highly relevant to the clinical symptoms and<br />

surgical procedures. 1 Recognition of the anatomical variants<br />

of the arterial blood supply to the AVN may help overcome<br />

potential complications following ablation procedures or in<br />

mitral valve surgery.<br />

Isolated right bundle branch block (RBBB) is a common<br />

finding in the general population. RBBB may be associated<br />

with structural heart disease but many subjects with this<br />

conduction abnormality have no evidence of underlying<br />

heart disease. 2, 3 The blood supply to the proximal part of<br />

the right bundle branch is provided by the left anterior<br />

descending artery (LAD) or the AVN artery, and the distal<br />

part is mainly supplied by branches from the LAD. 4<br />

With regard to the contribution of the AVN artery to the<br />

blood supply of the right bundle branch, we postulated that<br />

RBBB may be associated with anatomical variations in the<br />

AVN artery. The aim of this study was to investigate the<br />

origin of the AVN artery in patients with isolated RBBB and<br />

to compare it with normal electrocardiogram individuals.<br />

Methods<br />

This case control study was conducted enrolling patients<br />

with normal coronary arteries or mild coronary artery<br />

disease who underwent coronary angiography between 2001<br />

and 2010 in Tehran Heart Center. Demographic features,<br />

coronary risk factors, drug history, history of cardiac events,<br />

electrocardiographic interpretation, echocardiographic<br />

findings, past history of coronary or any other type of<br />

intervention and open heart surgery, and the result of<br />

coronary angiography were drawn out of the computerized<br />

angiography database of Tehran Heart Center. Patients eligible<br />

for inclusion were men and women at least 20 years of age<br />

with documented RBBB on the surface electrocardiogram,<br />

mild or normal coronary arteries proven by angiography, and<br />

a normal ejection fraction (more than 50%). Major exclusion<br />

criteria included the presence of conduction abnormalities<br />

other than RBBB, history of using drugs affecting the heart<br />

conduction system such as beta adrenergic receptor blockers<br />

and antiarrhythmic agents, significant valvular heart disease<br />

(more than mild regurgitation or stenosis of any valve) on<br />

echocardiography study, and congenital or acquired structural<br />

heart disease that could be an etiology for developing RBBB<br />

such as atrial septal defect, ventricular septal defect, and<br />

pulmonary thromboembolism. Case selection was done<br />

with convenience non-probability sampling. Initially, 141<br />

patients were selected from the angiography database.<br />

Subsequently, the hospital records of these patients,<br />

including the history of symptoms, further evaluation of<br />

the electrocardiogram, echocardiographic findings, and<br />

coronary angiography report sheets, were reviewed in detail.<br />

First, the electrocardiogram and records were selected for<br />

further study if the electrocardiogram satisfied the criteria<br />

previously defined for typical RBBB by the World Health<br />

Organization and the International Society and Federation<br />

for Cardiology in 1985, as follows: 1) prolongation of QRS<br />

to 0.12 second or more; 2) an rsr’, rsR’, or rSR’ pattern and<br />

occasionally a wide and notched R pattern in lead V 1<br />

or V 2<br />

;<br />

3) an S wave duration longer than the R wave duration or<br />

greater than 40 ms in leads V 6<br />

and I; and 4) an R peak time<br />

greater than 0.05 second in lead V 1<br />

but normal in leads V 5<br />

and V 6<br />

. 5<br />

Of these criteria, the first three should be present for the<br />

diagnosis to be made. When a notched dominant R pattern is<br />

present in V 1<br />

, criterion 4 should be satisfied as well.<br />

Of the initial 141 patients selected, 49 subjects who did<br />

not meet the inclusion criteria were excluded. Finally, a<br />

total of 92 subjects were entered for final analysis. Control<br />

subjects, who underwent coronary angiography during the<br />

same period, were randomly extracted from the angiography<br />

database. The controls were matched by age and sex to<br />

the cases with a 2:1 ratio. Inclusion criteria other than<br />

electrocardiography findings were the same as those of the<br />

cases. For each group, the coronary angiography recorded on<br />

CDs was studied to determine the pattern and characteristics<br />

of the coronary anatomy and the origination of the AVN<br />

artery. The dominant circulation was also determined.<br />

The classification described by Popma JJ was used in the<br />

present study to define the dominant coronary circulation 6 :<br />

a) in a right dominant circulation, the posterior descending<br />

artery (PDA) and at least one posterolateral branch (PLB)<br />

originate from the right coronary artery (RCA); b) in a left<br />

dominant circulation, the PDA and all of the PLBs originate<br />

from the left coronary artery; and c) In a co-dominant<br />

circulation, the PDA originates from the RCA and all of the<br />

PLBs originate from the left coronary artery.<br />

In order to determine the origin of the AVN artery, the<br />

dominant artery was ascertained. Based on the knowledge<br />

that the AVN artery most commonly originates from the<br />

crux, 7 if a branch to the AVN region originated from the<br />

dominant artery at the area of the crux, it was considered<br />

the AVN artery. If such a branch was not detected, other<br />

segments of the dominant artery and thereafter the nondominant<br />

artery were evaluated in multiple views. At the<br />

end, the origin of the AVN artery was classified to be from<br />

the right, left, or dual circulation (if both circulatory systems<br />

gave rise to the AVN blood supply). If the AVN artery was<br />

not originated from the crux, we determined the specific<br />

The Journal of Tehran University Heart Center165


The Journal of Tehran University Heart Center<br />

Ali Kazemisaeid et al.<br />

artery giving rise to it.<br />

Statistical analysis was performed using SPSS version<br />

15.0 software. Results for the quantitative variables were<br />

reported as mean ± standard deviation and as frequencies for<br />

the qualitative variables. The mean values of the quantitative<br />

variables of the two study groups were compared using the<br />

Student t-test. The frequency distributions of the qualitative<br />

variables, obtained from the two study groups, were<br />

compared using the chi-square test. A p value ≤ 0.05 was<br />

considered a statistically significant difference.<br />

Results<br />

A total of 276 patients were involved, and 92 (33.3%) and<br />

184 (66.7%) subjects were grouped as cases and controls,<br />

respectively. The mean age for the case group (59.8% male)<br />

was 59.19 ± 10.93 years and for the controls (62.5% male)<br />

59.29 ± 10.94 years. In terms of dominancy, there was no<br />

significant difference between the study groups (p value = 0.<br />

200). Also, 75.0% of the cases versus 76.6% of the controls<br />

had right dominant, 12.0% of the cases versus 16.3% of the<br />

controls had left dominant, and 13.0% of the cases versus<br />

7.1% of the controls had co-dominant circulations (Figure<br />

1). By comparison, in terms of dominancy, there was no<br />

significant difference between the study groups.<br />

Figure 1. Comparison between the cases (individuals with right bundle<br />

branch block) and controls (individuals without right bundle branch block),<br />

showing the frequency of the distribution of the types of dominancy<br />

In the evaluation of the AVN artery origin, we determined<br />

whether it was originated from the right or left or whether<br />

it had a dual origin, whether or not it was from the crux.<br />

Furthermore, we determined the specific artery, from which<br />

the AVN artery branched (Tables 1 and 2). The crux was<br />

defined as the U-turn segment of the dominant artery.<br />

Of the total study population, the AVN origin was from<br />

the crux in 106 (38.4%) and from a non-crux origin in 169<br />

(61.2%) subjects (Table 1).<br />

Table 1. Variations of the AVN origin artery in the total study population<br />

Origin of AVN artery<br />

Number of subjects<br />

Undetermined 1<br />

Crux 106<br />

Non-crux origins 169<br />

Dual origin<br />

PLB and distal of LCX 2<br />

OM and distal of RCA 1<br />

Origin from LCX<br />

Distal of LCX 3<br />

Mid-part of LCX 1<br />

Origin from OM 40<br />

Origin from RCA<br />

Conus 3<br />

Proximal of RCA 7<br />

Distal of RCA 24<br />

Posterior descending artery 2<br />

Posterolateral branch 85<br />

Origin from Ramus 1<br />

Total 276<br />

AVN, Atrioventricular node; LCX, Left circumflex artery; RCA, Right<br />

coronary artery; OM, Obtuse marginal artery<br />

In one patient, the AVN origin was undetermined, probably<br />

because it was too small to be visualized. Only a minority of<br />

the patients had an AVN artery with a dual origin (2 of the<br />

cases and one of the controls), as is shown in Table 1.<br />

These special cases were omitted from the subsequent<br />

analysis of the AVN origin because of the very small number<br />

of such cases. The results after such an adjustment are<br />

depicted in Table 2.<br />

Table 2 illustrates that origination of the AVN artery from<br />

a non-crux origin was more common than origination from<br />

the crux in both groups. The prevalence of the non-crux<br />

origination of the AVN artery was significantly higher in the<br />

cases than in the controls (79.8% vs. 51.4%, respectively; p<br />

value < 0.001).<br />

Although origination from the right circulatory system<br />

was more common in both groups (82.0% of the cases and<br />

68.9% of the controls), the prevalence of the right origin of<br />

the AVN artery was significantly higher in the cases than in<br />

the controls (p value = 0.021) (Table 2).<br />

A more detailed comparison of the AVN artery origin<br />

between the cases and controls is illustrated in Figure 2.<br />

The most common origin of the AVN artery was the crux<br />

in the controls and the PLB in the cases when considering<br />

the frequency of the different non-crux origin variations of<br />

the AVN artery separately. Nevertheless, by integrating these<br />

166


Anatomy of Atrioventricular Node Artery and Pattern of Dominancy in Normal ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

subtypes and considering the single variant of the “non-crux<br />

origin of the AVN artery”, it became more common than the<br />

crux origin even slightly in the controls.<br />

We analyzed the association between the origin of the<br />

AVN artery and the type of dominancy in the whole study<br />

population and observed that the AVN artery most commonly<br />

originated from the dominant artery. In the case of the codominant<br />

circulation, it most commonly originated from the<br />

right circulation (Table 3).<br />

Table 2. Comparison of the AVN artery origin in individuals with right<br />

bundle branch block (cases) and individuals without right bundle branch<br />

block )controls) *<br />

AVN artery<br />

origin<br />

Total<br />

n=272<br />

Case<br />

n=89<br />

Control<br />

n=183<br />

Left 73 (26.8) 16 (18.0) 57 (31.1)<br />

Right 199 (73.2) 73 (82.0) 126 (68.9)<br />

Crux 107 (39.3) 18 (20.2) 89 (48.6)<br />

Non-crux 165 (60.7) 71 (79.8) 94 (51.4)<br />

*<br />

Data are presented as n )%(<br />

**<br />

Case vs. Control<br />

AVN, Atrioventricular node<br />

P value **<br />

0.021<br />

< 0.001<br />

Figure 2. Frequency of the AVN artery origin in cases (individuals with<br />

right bundle branch block) and controls (individuals without right bundle<br />

branch block)<br />

AVN, Atrioventricular node; CX, Left circumflex artery; OM, Obtuse<br />

marginal artery; PDA, Posterior descending artery; PLB, Posterolateral<br />

branch artery; RCA, Right coronary artery<br />

Table 3. Association of the AVN artery origin with dominancy<br />

Dominancy<br />

Origin of the AVN artery<br />

from right circulation from left circulation<br />

Right dominant 87.9% 12.1%<br />

Left dominant 7.5% 92.5%<br />

Co-dominant 60.0% 40.0%<br />

AVN, Atrioventricular node<br />

Discussion<br />

The findings of the present study suggest that there is no<br />

relationship between the dominancy of the epicardial arteries<br />

and the presence of RBBB in subjects with normal coronary<br />

arteries. There was a great variation of the AVN artery origin<br />

among all the study subjects. In the total study population<br />

and also in each group, the AVN artery was more commonly<br />

separated from a non-crux origin than from the crux (61.2%<br />

versus 38.4%); this is in contrast to the conventional<br />

wisdom. 7 The prevalence of the non-crux origination of<br />

the AVN artery was significantly higher in the cases than<br />

in the controls. A dual artery supply of the AVN was a rare<br />

variation, only observed in 0.01% of the subjects. While<br />

the origination of the AVN artery from the right circulatory<br />

system was more common in both groups, the prevalence of<br />

the right origin of the AVN artery was significantly higher in<br />

the cases than in the controls. Finally, we observed that the<br />

AVN artery most commonly originated from the dominant<br />

artery but not necessarily from the crux.<br />

To this date, there has been no previous study aiming to<br />

specifically compare anatomic variations in the AVN artery<br />

in subjects with RBBB versus subjects with normal surface<br />

electrocardiograms.<br />

Table 4 represents the results of the previous studies that<br />

investigated the AVN artery origin. 8-14 All the studies showed<br />

that the AVN artery originated most commonly from the<br />

RCA, which was the most common dominant system. This<br />

led to the conclusion that the AVN artery usually branched<br />

from the dominant artery. However, this was not always the<br />

case as we observed some cases of the origination of the<br />

AVN artery from the non-dominant artery. These out-ofthe-rule<br />

variants were more commonly seen in the control<br />

group (15% of the controls versus 1% of the cases and 10%<br />

of the total study population). The results of the current<br />

study with regard to this variable were very close to the<br />

results of Ramanathan and his colleagues’ angiographybased<br />

study on an Indian population and somewhat different<br />

from other studies, suggesting that race might be a potential<br />

factor influencing the anatomic variables of the coronary<br />

artery. 14 Three per cent of the total population investigated<br />

by Ramanathan et al., showed origination of the AVN artery<br />

from the non-dominant artery.<br />

Pejković B et al., in an anatomic study of 150 human hearts,<br />

found that the artery supplying the AVN arose most frequently<br />

from the U- or V-shaped segment of the right (90%) or left<br />

(10%) coronary artery termed the ‘U-turn’ at the level of the<br />

crux. 15 Differently in our study, the AVN artery was more<br />

commonly separated from a non-crux origin than from the<br />

crux (61.2% versus 38.4%). In 72.1% of the subjects, it was<br />

a branch from the right circulation and in 26.4% from the left<br />

circulation. In line with our observation, Pejkovic B et al., also<br />

showed that the AVN artery origin depended on topographic<br />

The Journal of Tehran University Heart Center167


The Journal of Tehran University Heart Center<br />

Ali Kazemisaeid et al.<br />

arterial dominance: in cases with right or balanced arterial<br />

coronary vascularization, the AVN artery was a branch of the<br />

RCA, and in cases of left dominance the left coronary artery<br />

gave a branch to the AVN. 15<br />

Dual supply of the AVN was a rare variation in the present<br />

sample of individuals. Such a dual supply has been described<br />

in the literature previously. 8, 10, 12-14, 16, 17 The co-dominant<br />

circulation seems not to be related to the dual arterial blood<br />

supply of the AVN insofar as in our study none of the 3<br />

patients with dual supply were co-dominant (one was leftdominant<br />

and the other 2 were right-dominant).<br />

Rusu MC et al., in an anatomic study of 50 human hearts,<br />

described five morphological types (each with distinctive<br />

subtypes) of the AVN artery: 1) type I (22%, the AVN artery<br />

from the U-turn of the RCA); 2) type II (18%, the AVN artery<br />

from the PDA); 3) type III (34%, the AVN artery from the<br />

PLB); 4) type IV (8%, the AVN artery from the bifurcation of<br />

the RCA into the PDA and the PLB-trifurcated RCA); and 5)<br />

type V (18%, the AVN artery from the circumflex artery). 18<br />

According to this type of classification, in our total study<br />

population the prevalence of each subtype was as follows:<br />

38.4% type I; 0.7% type II; 30.8% type III; 8.6% type IV;<br />

and 1.4% type V. We also observed additional subtypes such<br />

as origination from the OM, conus, proximal part of the RCA<br />

and ramus in 14.5%, 1.1%, 2.5%, and 0.3% of the subjects,<br />

respectively. Similar to the Russu study, the separation of the<br />

AVN artery from a non-crux origin was more common than<br />

that from the crux. Also in both studies, the most common<br />

non-crux origin of the AVN artery was from the PLB. The<br />

least common subtype in our study was origination from<br />

the PDA (and also ramus), in contrast to the Russu study, in<br />

which origination from the PDA was a common variation.<br />

By comparing the two study groups in our investigation, as<br />

is shown in Figure 2, this conclusion could be readily made<br />

that the origin of the AVN artery differed in the subjects with<br />

RBBB and those without RBBB.<br />

Conclusion<br />

According to our observations, there was no relationship<br />

between the dominancy of the epicardial arteries and the<br />

presence of RBBB in subjects with normal coronary arteries.<br />

There was a great variation of the AVN artery origin. Noncrux<br />

origination of the AVN artery was more common than<br />

the crux origination in both groups, and the prevalence of<br />

non-crux origination of the AVN artery was significantly<br />

higher in the cases than in the controls. Origination of the<br />

AVN artery from the right circulatory system was more<br />

common in both groups and the prevalence of the right origin<br />

of the AVN artery was significantly higher in the cases than<br />

in the controls. The AVN artery most commonly originated<br />

from the dominant artery but not necessarily from the crux.<br />

Acknowledgment<br />

This study was approved and supported by Tehran<br />

University of Medical Sciences.<br />

References<br />

1. Abuin G, Nieponice A. New findings on the origin of the<br />

blood supply to the atrioventricular node. Clinical and surgical<br />

significance. Tex Heart Inst J 1998;25:113-117.<br />

2. Knilans TK. Right bundle branch block. In: Surawicz B, ed. Chou’s<br />

Electrocardiography In Clinical Practice. 6th ed. Philadelphia:<br />

Saunders; 2008. p. 99-100.<br />

3. Mirvis DM, Goldberger AL. Electrocardiography. In: Bonow RO,<br />

Mann DL, Zipes DP, Libby P, Braunwald E, eds. Braunwald’s<br />

Heart Disease, A Textbook of Cardiovascular Medicine. 9th ed.<br />

Philadelphia: Saunders; 2011. p. 146-147.<br />

4. Stambler BS, Rahimtoola SH, Ellenbogen KA. Pacing for<br />

atrioventricular conduction system disease. In: Ellenbogen<br />

KA, Kay GN, eds. Clinical Cardiac Pacing, Defibrillation, and<br />

Resynchronization Therapy. 3rd ed. Philadelphia: Saunders; 2007.<br />

p. 429-430.<br />

5. Willems JL, Robles de Medina EO, Bernard R, Coumel P, Fisch<br />

C, Krikler D, Mazur NA, Meijler FL, Mogensen L, Moret P.<br />

Criteria for intraventricular conduction disturbances and preexcitation.<br />

World Health Organizational/International Society and<br />

Federation for Cardiology Task Force Ad Hoc. J Am Coll Cardiol<br />

1985;5:1261-1275.<br />

6. Pompa JJ. Coronary arteriography. In: Bonow RO, Mann DL,<br />

Table 4. Origin of the AVN artery reported by various authors<br />

Author Study date Sample size<br />

Origin of AVN artery (%)<br />

RCA LCX Dual<br />

Vieweg et al. 1975 118 84 8 8<br />

Hutchison 1978 40 80 20 -<br />

Hadziselimović 1978 200 85 13 2<br />

Krupa 1993 120 90 10 -<br />

Futami et al. 2003 30 80 10 10<br />

Saremietal 2008 102 87 11 2<br />

Ramanathan et al. 2009 300 72 28 0<br />

The present study 2011 276 72.1 26.4 1.1<br />

AVN, Atrioventricular node; RCA, Right coronary artery; LCX, Left circumflex artery<br />

168


Anatomy of Atrioventricular Node Artery and Pattern of Dominancy in Normal ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Zipes DP, Libby P, Braunwald E, eds. Braunwald’s Heart Disease,<br />

A Textbook of Cardiovascular Medicine. 9th ed. Philadelphia:<br />

Saunders; 2011. p. 406-440.<br />

7. Rubart M, Zipes DP. Genesis of cardiac arrhythmias:<br />

electrophysiologic considerations. In: Bonow RO, Mann DL,<br />

Zipes DP, Libby P, Braunwald E, eds. Braunwald’s Heart Disease,<br />

A Textbook of Cardiovascular Medicine. 9th ed. Philadelphia:<br />

Saunders; 2011. p. 653-686.<br />

8. Vieweg WV, Alpert JS, Hagan AD. Origin of the sinoatrial node<br />

and atrioventricular node arteries in right, mixed, and left inferior<br />

emphasis systems. Cathet Cardiovasc Diagn 1975;1:361-373.<br />

9. Hutchinson MC. A study of the artrial arteries in man. J Anat<br />

1978;125:39-54.<br />

10. Hadziselimović H. Vascularization of the conducting system in the<br />

human heart. Acta Anat (Basel) 1978;102:105-110.<br />

11. Krupa U. The atrioventricular nodal artery in the human heart.<br />

Folia Morphol (Warsz) 1993;52:G1-9.<br />

12. Futami C, Tanuma K, Tanuma Y, Saito T. The arterial blood supply<br />

of the conducting system in normal human hearts. Surg Radiol<br />

Anat 2003;25:42-49.<br />

13. Saremi F, Abolhoda A, Ashikyan O, Milliken JC, Narula J,<br />

Gurudevan SV, Kaushal K, Raney A. Arterial supply to sinuatrial<br />

and atrioventricular nodes: imaging with multidetector CT.<br />

Radiology 2008;246:99-107.<br />

14. Ramanathan L, Shetty P, Nayak SR, Krishnamurthy A, Chettiar<br />

GK, Chockalingam A. Origin of the sinoatrial and atrioventricular<br />

nodal arteries in South Indians: an angiographic study. Arq Bras<br />

Cardiol 2009;92:314-319.<br />

15. Pejković B, Krajnc I, Anderhuber F, Kosutić D. Anatomical aspects<br />

of the arterial blood supply to the sinoatrial and atrioventricular<br />

nodes of the human heart. J Int Med Res 2008;36:691-698.<br />

16. Krauss D, Carter JE, Jr, Feldman T. Anomalous connection between<br />

the sinus node artery and the A-V node artery. Cathet Cardiovasc<br />

Diagn 1993;29:236-239.<br />

17. James TN, Burch GE. The atrial coronary arteries in man.<br />

Circulation 1958;17:90-98.<br />

18. Rusu MC, Ferechide D, Curca GC, Dermengiu D. Morph functional<br />

considerations on the atrioventricular node arterial vascularization.<br />

Rom J Leg Med 2009;17:101-110.<br />

The Journal of Tehran University Heart Center169


The Journal of Tehran University Heart Center<br />

Original Article<br />

Determinants of Length of Stay in Surgical Ward after<br />

Coronary Bypass Surgery: Glycosylated Hemoglobin as a<br />

Predictor in All Patients, Diabetic or Non-Diabetic<br />

Mahdi Najafi, MD * , Hamidreza Goodarzynejad, MD<br />

Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.<br />

Received 03 March 2012; Accepted 23 July 2012<br />

Abstract<br />

Background: Reports on the determinants of morbidity in coronary artery bypass graft surgery (CABG) have focused on<br />

outcome measures such as length of postoperative stay in the Intensive Care Unit (ICU). We proposed that major comorbidities<br />

in the ICU hampered the prognostic effect of other weaker but important preventable risk factors with effect on patients’<br />

length of hospitalization. So we aimed at evaluating postoperative length of stay in the ICU and surgical ward separately.<br />

Methods: We studied isolated CABG candidates who were not dialysis dependent. Preoperative, operative, and<br />

postoperative variables as well as all classic risk factors of coronary artery disease were recorded. Using multivariate<br />

analysis, we determined the independent predictors of length of stay in the ICU and in the surgical ward.<br />

Results: Independent predictors of extended length of stay in the surgical ward ( > 3 days) were a history of peripheral<br />

vascular disease, total administered insulin during a 24-hour period after surgery, glycosylated hemoglobin (HbA1c), last<br />

fasting blood sugar of the patients before surgery, and inotropic usage after cardiopulmonary bypass. The area under the<br />

Receiver Operating Characteristic Curve (AUC) was found to be 0.71 and Hosmer-Lemeshow (HL) goodness of fit statistic p<br />

value was 0.88. Independent predictors of extended length of stay in the ICU ( > 48 hours) were surgeon category, New York<br />

Heart Association functional class, intra-aortic balloon pump, postoperative arrhythmias, total administered insulin during<br />

a 24-hour period after surgery, and mean base excess of the first 6 postoperative hours (AUC = 0.70, HL p value = 0.94 ).<br />

Conclusion: This study revealed that the indices of glycemic control were the most important predictors of length of stay in<br />

the ward after cardiac surgery in all patients, diabetic or non-diabetic. However, because HbA1c level did not change under<br />

the influence of perioperative events, it could be deemed a valuable measure in predicting outcome in CABG candidates.<br />

J Teh Univ Heart Ctr 2012;7(4):170-176<br />

This paper should be cited as: Najafi M, Goodarzynejad H. Determinants of Length of Stay in Surgical Ward after Coronary Bypass<br />

Surgery: Glycosylated Hemoglobin as a Predictor in All Patients, Diabetic or Non-Diabetic. J Teh Univ Heart Ctr 2012;7(4):170-176.<br />

Keywords: Coronary artery bypass • Treatment outcome • Hemoglobin A, glycosylated • Length of stay<br />

Introduction<br />

In a world of limited medical care resources, despite<br />

increasing demand for medical services, clinicians often<br />

need to identify patients that are likely to require a longer<br />

period of high dependency care after cardiac surgery.<br />

Coronary artery bypass graft surgery (CABG) has received<br />

particular attention as it is an expensive and commonly<br />

*<br />

Corresponding Author: Mahdi Najafi, Assistant Professor of Anesthesiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran<br />

Heart Center, North Karegar Street, Tehran, Iran. 1411713138. Tel: +98 21 88029600. Fax: +98 21 88029731. E-mail: najafik@sina.tums.ac.ir.<br />

170


Determinants of Length of Stay in Surgical Ward after Coronary Bypass Surgery ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

performed procedure. 1, 2 Indeed, detection of patients who<br />

are likely to need a longer postoperative length of stay (LOS)<br />

for CABG would allow operations on these patients to be<br />

spread over the operating week. This would maximize the<br />

chance of the availability of the Intensive Care Unit (ICU)<br />

beds for other sick patients and also prevent the cancellations<br />

of operations. Moreover, prolonged ICU and ward stay in<br />

patients undergoing CABG increases overall hospital costs. 3<br />

LOS in hospital after cardiac surgery is among the most<br />

important determinants of outcome to have been studied<br />

so far. Many studies have reported the factors affecting the<br />

length of postoperative ICU stay. 4-12 A few investigators have<br />

evaluated total hospital LOS, 13-16 but to our knowledge, no<br />

study has assessed such predictors at cardiac surgical wards.<br />

Although the total time of hospital stay contains the time<br />

of stay in the ward, prolonged stay in the surgical ward is<br />

important itself inasmuch as it limits the number of available<br />

ICU beds because of a reduced throughput in the surgical<br />

ward, and as a consequence in the ICU unit. We proposed<br />

that the predictors of a prolonged ICU stay might be different<br />

from the predictors of a long postoperative stay in the<br />

hospital ward.<br />

Methods<br />

We prospectively studied a total of 570 consecutive patients<br />

undergoing elective CABG at Tehran Heart Center during<br />

a period of five months. Patients who underwent CABG<br />

combined with a heart valve repair or replacement, resection<br />

of a ventricular aneurysm, or other surgical procedures<br />

were excluded. The study was approved by the division of<br />

cardiothoracic surgery and by the research department of the<br />

hospital.<br />

Data were collected prospectively during the patients’<br />

admission by interviews and physical examinations on the<br />

following variables: age; gender; body mass index (BMI);<br />

educational level; New York Heart Association (NYHA)<br />

functional class; number of diseased vessels; and left<br />

ventricular ejection fraction (LVEF). A history of myocardial<br />

infarction, smoking, alcohol abuse, opiate dependency,<br />

diabetes, hypercholesterolemia, hypertension, peripheral<br />

vascular disease, cerebrovascular disease, respiratory failure,<br />

and renal failure were also noted.<br />

Laboratory data included measured blood sugar in<br />

perioperative and glycated or glycosylated hemoglobin<br />

(HbA1c) at operation day.<br />

The patients’ data, risk factors, operation, and outcome<br />

data were recorded in a structured form. Perioperative<br />

medical characteristics were collected by research general<br />

practitioners. The data were transcribed onto the SPSS<br />

software by a data entry operator at a later date.<br />

Regarding educational level, three levels of education<br />

were categorized: 1 = primary school or lower; 2 =<br />

secondary school; and 3 = university/college or equivalents.<br />

Current smoker was defined as anybody who had smoked<br />

within one month of surgery. Alcohol abuse was defined<br />

as the consumption of alcohol despite recurrent adverse<br />

consequences. Daily regular use of opium products was<br />

defined as opium dependency according to the DSM-IV<br />

criteria. (American Psychiatric Association. Diagnostic<br />

and statistical manual of mental disorders: DSM-IV-TR.<br />

American Psychiatric Publishing, Inc.; 2000) Any cerebral<br />

neurological deficit induced by both cerebrovascular<br />

accident and transient ischemic attacks or previous cerebral<br />

surgery was defined as cerebrovascular disease. Peripheral<br />

vascular disease was defined as a history or any evidence<br />

of aneurysm or occlusive peripheral vascular disease on<br />

physical examination. After surgery, the patients who stayed<br />

in the ward for more than 2 consecutive days on the initial<br />

admission were classified as having a prolonged ward stay.<br />

A long cardiac surgical ICU stay was defined as > 2 days.<br />

The patients within the first 24 hours after bypass surgery<br />

were categorized into three groups according to the average<br />

amount of insulin intake: group 0 = zero U; group 1 = 1 to<br />

9 U; and group 2 = 10 or more U of insulin. The operations<br />

were performed by seven surgeons. The surgeons, in terms<br />

of mean total in-hospital duration of stay for the patients of<br />

each individual surgeon, were classified into 3 groups, as<br />

follows: 1 = total LOS < 8 days; 2 = 8 ≤ total LOS < 10<br />

days; and 3 = total LOS ≥ 10 days. 17 Central and peripheral<br />

circulations were measured by the amount of inotropic<br />

support and arterial blood gas parameters (base excess and pH<br />

in plasma), respectively. A wound infection was any wound<br />

infection in the sternum or leg incision following surgery.<br />

Postoperative arrhythmias were all observed rhythms that<br />

needed treatment rather than normal sinus rhythm.<br />

The numerical variables are presented as mean ± SD,<br />

while the categorized variables are summarized by absolute<br />

frequencies and percentages. The continuous variables<br />

were compared using the Student t-test, and the categorical<br />

variables were compared using the chi-square (or the<br />

Fisher exact test, as required) and the Mantel-Haenszel<br />

chi-square test for trend. A multivariate forward stepwise<br />

logistic regression model for risk factors predicting LOS<br />

in the ICU and the surgical ward was constructed. Through<br />

the multivariate forward stepwise analysis, the variables<br />

were entered into the logistic regression model according<br />

to their statistical significance (entering criterion p value ≤<br />

0.15). The associations between the independent predictors<br />

and LOS in the ICU or the surgical ward in the final model<br />

were expressed as odds ratios (OR) with 95% CIs. Model<br />

discrimination was measured using the c statistic, which<br />

is equal to the area under the ROC (Receiver Operating<br />

Characteristic) curve. Model calibration was estimated<br />

using the Hosmer-Lemeshow (HL) goodness-of-fit statistic<br />

(higher p values imply that the model fits the observed data<br />

better). For the statistical analyses, the statistical software<br />

The Journal of Tehran University Heart Center171


The Journal of Tehran University Heart Center<br />

Mahdi Najafi et al.<br />

SPSS version 13.0 for Windows (SPSS Inc., Chicago, IL)<br />

and the statistical package SAS version 9.1 for Windows<br />

(SAS Institute Inc., Cary, NC, USA) were used. All the p<br />

values were two-tailed, with statistical significance defined<br />

by p value ≤ 0.05.<br />

Results<br />

The baseline characteristics, preoperative data, and<br />

outcome variables of the CABG patients according to LOS in<br />

the ICU are presented in Table 1. Overall, 380/570 (66.7%)<br />

patients were discharged from the ICU in ≤ 48 hours, and 190<br />

(33.3%) patients spent > 48 hours in the ICU. Table 2 shows<br />

the predictors of LOS in the ICU in the patients, undergoing<br />

bypass surgery. Three most important variables were intraaortic<br />

balloon pump support, mean 24 hours’ insulin intake<br />

of ≥ 10 units, and NYHA functional class, with odds ratios<br />

of 25.5, 2.3, and 2.2, respectively.<br />

The relationship between the LOS in the ICU and the<br />

Table 1. Baseline characteristics of the coronary artery bypass graft patients according to length of stay in the Intensive Care Unit<br />

ICU stay ≤ 48 hours (n=380) ICU stay > 48 hours (n=190) P value<br />

Age (y) 58.44±8.80 60.21±9.20 0.026<br />

Male sex 290 (76.3) 139 (73.2) 0.410<br />

Body mass index (kg/m 2 ) 27.14±4.09 27.84±4.01 0.055<br />

Surgeon category 0.017<br />

1 218 (57.4) 102 (53.7)<br />

2 104 (27.4) 41 (21.6)<br />

3 58 (15.3) 47 (24.7)<br />

Cardiac risk factors<br />

Diabetes mellitus 159 (41.8) 74 (38.9) 0.508<br />

Hypertension 178 (46.8) 104 (54.7) 0.076<br />

Hyperlipidemia 264 (69.5) 136 (71.6) 0.605<br />

Cigarette smoking 138 (36.3) 68 (35.8) 0.902<br />

Family history of CAD 186 (48.9) 84 (44.2) 0.286<br />

Prior MI 187 (49.5) 100 (52.9) 0.440<br />

Prior CVA 10 (2.6) 12 (6.3) 0.031<br />

History of PVD 100 (26.3) 58 (30.5) 0.290<br />

NYHA functional class 0.032<br />

1 141 (37.1) 60 (31.6)<br />

2 193 (50.8) 93 (48.9)<br />

3 46 (12.1) 37 (19.5)<br />

LVEF (%) 48.88±9.80 47.78±11.38 0.253<br />

Number of diseased vessels 0.607<br />

1 12 (3.2) 9 (4.7)<br />

2 73 (19.2) 35 (18.4)<br />

3 295 (77.6) 146 (76.8)<br />

Operative details and complications<br />

Pump time (min) 68.45±20.36 73.19±25.24 0.025<br />

Number of grafts 3.70±0.94 3.79±0.99 0.265<br />

Intraoperative IABP-support 1 (0.3) 11 (5.8) < 0.001<br />

Endarterectomy 27 (7.1) 12 (6.3) 0.725<br />

Pacemaker dependents 33 (8.7) 20 (10.5) 0.475<br />

Inotropic drug use 148 (38.9) 90 (47.4) 0.055<br />

Arrhythmia 123 (32.4) 101 (53.2) < 0.001<br />

Perioperative MI 1 (0.3) 3 (1.6) 0.110<br />

Respiratory failure 56 (14.7) 48 (25.3) 0.002<br />

Wound infection 0 3 (1.6) 0.037<br />

Arterial blood gas parameters<br />

Mean BE within 6 hours after surgery (mmol/l) -5.42±2.55 -6.19±2.57 0.001<br />

pH < 7.34 within 6 hours after surgery 74 (19.5) 59 (31.1) 0.002<br />

Mean insulin intake within 24 hours after surgery < 0.001<br />

Zero U 193 (50.8) 69 (36.3)<br />

1 - 9 U 99 (26.1) 48 (25.3)<br />

≥ 10 U 88 (23.2) 73 (38.4)<br />

*<br />

Data are presented as mean±SD or n (%)<br />

ICU, Intensive Care Unit; CAD, Coronary artery disease; MI, Myocardial infarction; CVA, Cerebrovascular accident; PVD, Peripheral vascular disease;<br />

NYHA, New York Heart Association; LVEF, Left ventricular ejection fraction; IABP, Intra-aortic balloon pump; BE, Base excess<br />

172


Determinants of Length of Stay in Surgical Ward after Coronary Bypass Surgery ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Table 2. Predictors of length of stay in the Intensive Care Unit in patients undergoing bypass surgery<br />

Predictors<br />

Univariate analysis<br />

Multivariate analysis<br />

OR<br />

95% CI<br />

P value<br />

OR<br />

95% CI<br />

P value<br />

NYHA classification<br />

0.05<br />

0.02<br />

2 vs. 1<br />

1.13<br />

0.76-1.67<br />

1.17 0.77-1.79<br />

3 vs. 1<br />

1.89<br />

1.11-3.20<br />

2.15 1.22-3.79<br />

Surgeon category<br />

0.02<br />

0.04<br />

Group 2 vs. 1<br />

0.84<br />

0.54-1.29<br />

0.69 0.43-1.12<br />

Group 3 vs. 1<br />

1.73<br />

1.10-2.71<br />

1.47 0.91-2.38<br />

Intra-aortic balloon pump<br />

support<br />

23.29<br />

2.98-181.79 < 0.01 25.22 3.14-202.43 < 0.01<br />

Postoperative arrhythmia<br />

2.37<br />

1.65-3.38<br />

< 0.01<br />

1.95 1.33-2.86<br />

< 0.01<br />

Mean BE within 6 hours<br />

after surgery<br />

0.88<br />

0.82-0.95<br />

< 0.01<br />

0.90 0.83-0.97<br />

< 0.01<br />

Mean insulin intake within<br />

24 hours after surgery *<br />

1 vs. 0<br />

1.35<br />

0.87-2.10<br />

< 0.01<br />

1.31 0.82-2.09<br />

< 0.01<br />

2 vs. 0<br />

*<br />

Group 0 = Zero U<br />

Group 1 = 1 to 9 U<br />

Group 2 = ≥ 10 U<br />

2.32<br />

1.53-3.51<br />

2.27 1.45-3.55<br />

OR, Odds ratio; CI, Confidence interval; NYHA, New York Heart Association; BE, Base excess; AUC, Area under the Receiver Operating Characteristic<br />

Curve (c = 0.70)<br />

Table 3. Durations of Intensive Care Unit, surgical ward and total hospital stay as well as in-hospital mortality rate for both study groups *<br />

Group A<br />

Group B<br />

Patients with ICU stay ≤ 48h (n=380)<br />

Patients with ICU stay > 48h (n=190)<br />

P value<br />

ICU stay (hr) 27.38±9.30 85.79±34.87<br />

Surgical ward stay (d) 2.78±1.79 2.63±2.35 0.43<br />

Total hospital stay (d) 7.21±2.84 9.85±7.36 < 0.01<br />

In-hospital mortality 0 3 (1.6) 0.04<br />

*<br />

Data are presented as mean±SD or n (%)<br />

Table 4. Comparison of patients with regard to LOS in ward after CABG *<br />

Ward stay ≤ 3 days (n=426) Ward stay > 3 days (n=144) P value<br />

Male sex<br />

336 (78.9) 93 (64.6) < 0.01<br />

Body mass index (kg/m 2 ) 27.13±3.96 28.12±4.31 0.01<br />

Education class ** < 0.01<br />

1 218 (51.2) 94 (65.3)<br />

2 133 (31.2) 36 (25.0)<br />

3 75 (17.6) 14 (9.7)<br />

Diabetes mellitus 153 (35.9) 80 (55.6) < 0.01<br />

Family history of CAD 191 (44.8) 79 (54.9) 0.04<br />

History of PVD 104 (24.4) 54 (37.5) < 0.01<br />

Inotropic drug use 166 (39.0) 72 (50.0) 0.02<br />

Postoperative arrhythmia 179 (42.0) 45 (31.3) 0.02<br />

HbA1C at operation day 5.83±1.58 6.55±1.90 < 0.01<br />

Last fasting blood sugar 103.58±30.41 119.45±46.88 < 0.01<br />

Mean insulin intake within 24 hours after surgery *** < 0.01<br />

0 220 (51.6) 42 (29.2)<br />

1 100 (23.5) 47 (32.6)<br />

2 106 (24.9) 55 (38.2)<br />

Mean BG within the first 24 hours after surgery (mg/dl) 167.41±26.28 176.49±28.10 < 0.01<br />

Blood urea nitrogen (mg/dl) 38.86±11.56 42.19±12.92 < 0.01<br />

Total cholesterol (mg/dl) 157.97±43.89 169.19±47.32 < 0.01<br />

*<br />

Data are presented as mean±SD or n (%)<br />

**<br />

1 = primary school or lower; 2 = secondary school; 3 = university/college or equivalents<br />

***<br />

Group 0 = Zero U; Group 1 = 1 to 9 U; Group 2 ≥ 10 U<br />

LOS, Length of stay; CABG, Coronary artery bypass surgery; CAD, Coronary artery disease; PVD, Peripheral vascular disease; HbA1C, Glycosylated<br />

hemoglobin; BG, Blood glucose<br />

The Journal of Tehran University Heart Center173


The Journal of Tehran University Heart Center<br />

Mahdi Najafi et al.<br />

Table 5. Predictors of length of stay at surgical ward in patients undergoing bypass surgery<br />

Univariate analysis<br />

Multivariate analysis<br />

OR 95% CI P value OR 95% CI P value<br />

Peripheral vascular disease 1.858 1.241-2.781 < 0.01 1.568 1.013-2.426 0.04<br />

HbA1c 1.268 1.133-1.419 < 0.01 1.210 1.074-1.362 < 0.01<br />

Last fasting blood sugar 1.011 1.006-1.016 < 0.01 1.009 1.003-1.014 < 0.01<br />

Inotropic drug use 1.566 1.071-2.291 0.02 1.638 1.088-2.465 0.02<br />

Postoperative arrhythmia 0.627 0.420-0.937 0.02 0.531 0.343-0.822 0.01<br />

Mean insulin intake within 24 hours after surgery * < 0.01 < 0.01<br />

1 vs. 0 2.462 1.525-3.973 2.455 1.487-4.054<br />

2 vs. 0 2.718 1.709-4.321 2.708 1.648-4.450<br />

*<br />

Group 0 = Zero U<br />

Group 1 = 1 to 9 U<br />

Group 2 ≥ 10 U<br />

OR, Odds ratio; CI, Confidence interval; NYHA, New York Heart Association; HbA1c, Glycosylated hemoglobin<br />

surgical ward was investigated through a comparison of the<br />

average ICU, surgical ward, and total hospital stay as well<br />

as in-hospital mortality rate between two groups of ICU stay<br />

( ≤ 48 and > 48 hours) as listed in Table 3. The overall inhospital<br />

mortality rate was 0.5%.<br />

In all the patients studied, the LOS at the cardiac surgical<br />

ward ranged from 1 to 14 days (mean ± SD, 2.73 ± 1.99<br />

days) with a median of 2 days. Patients with a prolonged<br />

surgical ward stay (144/570, 25.3%) were more likely to be<br />

women and have diabetes, family history of coronary artery<br />

disease, history of peripheral disease, and lower educational<br />

level.<br />

Table 4 compares the patients’ characteristics by taking<br />

into consideration the LOS in the ward after cardiac surgery.<br />

Among the important factors that were significantly different<br />

between the two groups, i.e. LOS > 3 days and LOS ≤ 3 days,<br />

five factors were independent predictors. Three predictors<br />

were indices of glycemic control (Table 5). The area under<br />

the Receiver Operating Characteristic Curve (AUC) was<br />

found to be 0.71, which showed a good predictive accuracy<br />

in the variables included in the model. The model was also<br />

found to fit the data well since the Hosmer-Lemeshow p<br />

value was 0.88 (p < 0.05 indicated a poor fit).<br />

Discussion<br />

In the current study, we included preoperative,<br />

intraoperative, and immediate postoperative variables to<br />

find LOS determinants in 570 CABG patients after bypass<br />

surgery. The six variables of intra-aortic balloon pump<br />

(IABP), NYHA functional class, postoperative arrhythmia,<br />

24-hour average insulin intake, mean 6-hour BE, and surgeon<br />

category were found to be the independent predictors of<br />

an ICU LOS greater than 48 hours. We also found five<br />

predictors to be the independent risk factors for an increased<br />

cardiac surgical ward LOS. Considerable reports have been<br />

published on the predictors of LOS in the ICU following<br />

CABG 4-12 and some studies have investigated factors<br />

increasing postoperative hospital stay. 13-16 To our knowledge,<br />

however, no study has focused on the determinants of the<br />

LOS of cardiac surgical wards. Thus, we think that our study<br />

may contribute to the literature in this regard.<br />

Over the past two decades, attempts have been made to find<br />

the determinants of postoperative LOS in CABG patients.<br />

Be that as it may, a great disparity in type and number of<br />

independent variables analyzed has been reported. Although<br />

most of outcome studies with the focus on LOS have<br />

investigated only preoperative and intraoperative variables,<br />

there are some outcome research reports with immediate<br />

6, 13<br />

postoperative variables included.<br />

Poor cardiac output states, as reflected by the need for<br />

an IABP and inotropic therapy, have been identified as a<br />

predictor of a prolonged ICU stay in several studies. 6, 8, 9 In<br />

the present study, use of IABP was an ICU stay predictor,<br />

whereas inotropic support was a surgical ward stay predictor.<br />

Additionally, both reflected cardiac output.<br />

Need for an IABP was identified as an independent<br />

risk factor for ICU stay longer than 48 hours in three<br />

studies. 18-20 In contrast, Doering et al. showed that only early<br />

hemodynamic instability was indicative of an ICU stay<br />

longer than 24 hours, whereas postoperative IABP and use<br />

of inotropic drugs were not. 9 Michalopoulos et al. also noted<br />

that the number of inotropes, rather than inotrope use itself,<br />

was the significant factor. 6<br />

These differences may result from different treatment<br />

protocols or the way in which variables such as the use of<br />

inotropes were defined. Michaloupolous et al. used a 6- hour<br />

window for starting inotropic support after surgery and did<br />

not consider vasodilators. 6 By contrast, in the Doering et al.<br />

study, a 3-hour period was adopted and after-load reducers<br />

such as nitroprusside were considered in the definition of<br />

inotropic support. 9 Christakis et al. did not consider inotropic<br />

support alone, adding to IABP support in defining low output<br />

state. 7<br />

Some disparities in study results may be due to various<br />

study populations and treatment protocols, different cut-offs<br />

used to define a prolonged stay in the ICU (from ≥ 2 to 10<br />

174


Determinants of Length of Stay in Surgical Ward after Coronary Bypass Surgery ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

days), 5, 14, 21-23 differences in variables definitions, and the<br />

large number of variables that could be considered. 4<br />

The main difference between our study and those described<br />

above is that we took into account surgeon category and<br />

amount of insulin administered during a 24-hour period after<br />

CABG. We found that a patient’s surgeon could potentially<br />

influence the duration of ICU stay and that there were<br />

differences in the average length of ICU stay between the<br />

surgeons at our hospital. 17 It may indicate the necessity of<br />

using a unique protocol for discharging the patients from<br />

the ICU and hospital after surgery. We also found that the<br />

LOS in the ICU and ward depended predominantly on the<br />

amount of insulin received by the patients during the first 24-<br />

hour postoperative period. In addition, we could not show<br />

any association between the LOS in the ICU and that in the<br />

surgical ward (Table 3). As a result, a prolonged stay in the<br />

ICU probably is not an independent predictor of surgical<br />

ward stay.<br />

We found that the blood glucose control indices, including<br />

HbA1c and last fasting blood sugar (FBS) before surgery,<br />

were the risk factors for an extended LOS in surgical wards<br />

in patients undergoing CABG, diabetic or non-diabetic.<br />

Finding both HbA1c and FBS as the determinants of<br />

the LOS in the surgical ward suggests that HbA1c is an<br />

independent predictor of hospital stay regardless of the level<br />

of blood sugar. However, as HbA1c is an index of glycemic<br />

control during previous months, its level is independent of<br />

perioperative events. It may, therefore, be a more stable<br />

and reliable perioperative marker. HbA1c has been used<br />

as a marker of short-term glycemic control in diabetic<br />

patients for many years. Recently, the American Diabetes<br />

Association included HbA1c in diabetes mellitus diagnosis<br />

criteria. 24 The role of HbA1c in predicting morbidity and<br />

mortality in non-diabetic healthy people has been suggested<br />

by some population-based studies. 25, 26 However, there is<br />

limited knowledge about the prognostic role of HbA1c for<br />

perioperative outcome, and the relationship between HbA1c<br />

and postoperative outcome in cardiovascular surgery is<br />

controversial. 27-30<br />

Our study revealed that HbA1c could predict postoperative<br />

LOS in the surgical ward and not in the ICU. This may be<br />

explained by the hampering effect of more important strong<br />

cofactors such as IABP and NYHA functional class.<br />

Conclusion<br />

Among the five predictors of the LOS in the surgical<br />

ward, three were the indices of glycemic control. HbA1c is<br />

the most reliable marker of outcome because its level is not<br />

influenced by perioperative events. Taking these findings<br />

into consideration, the prognostic role of HbA1c for the<br />

LOS following CABG is promising. We would, therefore,<br />

recommend its use as a simple predictor of outcome after<br />

cardiac surgery in daily practice.<br />

Acknowledgments<br />

This study was approved and supported by Tehran<br />

University of Medical Sciences.<br />

References<br />

1. Nilsson J, Algotsson L, Höglund P, Lührs C, Brandt J. EuroSCORE<br />

predicts intensive care unit stay and costs of open heart surgery.<br />

Ann Thorac Surg 2004;78:1528-1534.<br />

2. Riordan CJ, Engoren M, Zacharias A, Schwann TA, Parenteau GL,<br />

Durham SJ, Habib RH. Resource utilization in coronary artery<br />

bypass operation: does surgical risk predict cost Ann Thorac Surg<br />

2000;69:1092-1097.<br />

3. Doering LV, Esmailian F, Laks H. Perioperative predictors of ICU<br />

and hospital costs in coronary artery bypass graft surgery. Chest<br />

2000;118:736-743.<br />

4. Tuman KJ, McCarthy RJ, March RJ, Najafi H, Ivankovich AD.<br />

Morbidity and duration of ICU stay after cardiac surgery. A model<br />

for preoperative risk assessment. Chest 1992;102:36-44.<br />

5. Tu JV, Mazer CD, Levinton C, Armstrong PW, Naylor CD. A<br />

predictive index for length of stay in the intensive care unit<br />

following cardiac surgery. CMAJ 1994;151:177-185.<br />

6. Michalopoulos A, Tzelepis G, Pavlides G, Kriaras J, Dafni U,<br />

Geroulanos S. Determinants of duration of ICU stay after coronary<br />

artery bypass graft surgery. Br J Anaesth 1996;77:208-212.<br />

7. Christakis GT, Fremes SE, Naylor CD, Chen E, Rao V, Goldman<br />

BS. Impact of preoperative risk and perioperative morbidity on<br />

ICU stay following coronary bypass surgery. Cardiovasc Surg<br />

1996;4:29-35.<br />

8. Ryan TA, Rady MY, Bashour CA, Leventhal M, Lytle B, Starr NJ.<br />

Predictors of outcome in cardiac surgical patients with prolonged<br />

intensive care stay. Chest 1997;112:1035-1042.<br />

9. Doering LV, Esmailian F, Imperial-Perez F, Monsein S.<br />

Determinants of intensive care unit length of stay after coronary<br />

artery bypass graft surgery. Heart Lung 2001;30:9-17.<br />

10. Janssen DP, Noyez L, Wouters C, Brouwer RM. Preoperative<br />

prediction of prolonged stay in the intensive care unit for coronary<br />

bypass surgery. Eur J Cardiothorac Surg 2004;25:203-207.<br />

11. Ghotkar SV, Grayson AD, Fabri BM, Dihmis WC, Pullan DM.<br />

Preoperative calculation of risk for prolonged intensive care unit<br />

stay following coronary artery bypass grafting. J Cardiothorac<br />

Surg 2006;1:14.<br />

12. Rosenfeld R, Smith JM, Woods SE, Engel AM. Predictors and<br />

outcomes of extended intensive care unit length of stay in patients<br />

undergoing coronary artery bypass graft surgery. J Card Surg<br />

2006;21:146-150.<br />

13. Weintraub WS, Jones EL, Craver J, Guyton R, Cohen C.<br />

Determinants of prolonged length of hospital stay after coronary<br />

bypass surgery. Circulation 1989;80:276-284.<br />

14. Mounsey JP, Griffith MJ, Heaviside DW, Brown AH, Reid DS.<br />

Determinants of the length of stay in intensive care and in hospital<br />

after coronary artery surgery. Br Heart J 1995;73:92-98.<br />

15. Lazar HL, Fitzgerald C, Gross S, Heeren T, Aldea GS, Shemin RJ.<br />

Determinants of length of stay after coronary artery bypass graft<br />

surgery. Circulation 1995;92:II20-24.<br />

16. Jensen L, Yang L. Risk factors for postoperative pulmonary<br />

complications in coronary artery bypass graft surgery patients. Eur<br />

J Cardiovasc Nurs 2007;6:241-246.<br />

17. Najafi M, Goodarzinejad H, Sheikhfathollahi M, Adibi H. Role of<br />

surgeon in length of stay in ICU after cardiac bypass surgery. J Teh<br />

Univ Heart Ctr 2010;1:9-13.<br />

18. Wong DT, Cheng DC, Kustra R, Tibshirani R, Karski J, Carroll-<br />

The Journal of Tehran University Heart Center175


The Journal of Tehran University Heart Center<br />

Mahdi Najafi et al.<br />

Munro J, Sandler A. Risk factors of delayed extubation, prolonged<br />

length of stay in the intensive care unit, and mortality in patients<br />

undergoing coronary artery bypass graft with fast-track cardiac<br />

anesthesia: a new cardiac risk score. Anesthesiology 1999;91:936-<br />

944.<br />

19. Bucerius J, Gummert JF, Walther T, Doll N, Falk V, Schmitt DV,<br />

Mohr FW. Predictors of prolonged ICU stay after on-pump versus<br />

off-pump coronary artery bypass grafting. Intensive Care Med<br />

2004;30:88-95.<br />

20. Hein OV, Birnbaum J, Wernecke K, England M, Konertz W, Spies<br />

C. Prolonged intensive care unit stay in cardiac surgery: risk factors<br />

and long-term-survival. Ann Thorac Surg 2006;81:880-885.<br />

21. Staat P, Cucherat M, George M, Lehot JJ, Jegaden O, André-Fouët<br />

X, Beaune J. Severe morbidity after coronary artery surgery:<br />

development and validation of a simple predictive clinical score.<br />

Eur Heart J 1999;20:960-966.<br />

22. Kurki TS, Kataja M. Preoperative prediction of postoperative<br />

morbidity in coronary artery bypass grafting. Ann Thorac Surg<br />

1996;61:1740-1745.<br />

23. Miller KH. Factors influencing selected lengths of ICU stay for<br />

coronary artery bypass patients. J Cardiovasc Nurs 1998;12:52-61.<br />

24. American Diabetes Association. Diagnosis and classification of<br />

diabetes mellitus. Diabetes Care 2010;33:S62-69.<br />

25. Silbernagel G, Grammer TB, Winkelmann BR, Boehm BO, März<br />

W. Glycated hemoglobin predicts all-cause, cardiovascular, and<br />

cancer mortality in people without a history of diabetes undergoing<br />

coronary angiography. Diabetes Care 2011;34:1355-1361.<br />

26. Selvin E, Steffes MW, Zhu H, Matsushita K, Wagenknecht L,<br />

Pankow J, Coresh J, Brancati FL. Glycated hemoglobin, diabetes,<br />

and cardiovascular risk in nondiabetic adults. N Engl J Med<br />

2010;362:800-811.<br />

27. Liu Y, Yang YM, Zhu J, Tan HQ, Liang Y, Li JD. Prognostic<br />

significance of hemoglobin A1c level in patients hospitalized with<br />

coronary artery disease. A systematic review and meta-analysis.<br />

Cardiovasc Diabetol 2011;10:98.<br />

28. Medhi M, Marshall MC, Jr, Burke HB, Hasan R, Nayak D, Reed G,<br />

LaFaro R, Southren AL. HbA1c predicts length of stay in patients<br />

admitted for coronary artery bypass surgery. Heart Dis 2001;3:77-<br />

79.<br />

29. O’Sullivan CJ, Hynes N, Mahendran B, Andrews EJ, Avalos G,<br />

Tawfik S, Lowery A, Sultan S. Haemoglobin A1c (HbA1C) in nondiabetic<br />

and diabetic vascular patients. Is HbA1C an independent<br />

risk factor and predictor of adverse outcome Eur J Vasc Endovasc<br />

Surg 2006;32:188-197.<br />

30. Goode KM, John J, Rigby AS, Kilpatrick ES, Atkin SL, Bragadeesh<br />

T, Clark AL, Cleland JG. Elevated glycated haemoglobin is a<br />

strong predictor of mortality in patients with left ventricular<br />

systolic dysfunction who are not receiving treatment for diabetes<br />

mellitus. Heart 2009;95:917-923.<br />

176


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Original Article<br />

Obvious or Subclinical Right Ventricular Dysfunction in<br />

Diabetes Mellitus (Type II): An Echocardiographic Tissue<br />

Deformation Study<br />

Mozhgan Parsaee, MD * , Parvaneh Bahmanziari, MD, Maryam Ardeshiri, MD,<br />

Maryam Esmaeilzadeh, MD<br />

Rajaei Cardiovascular, Medical and Research Center, Tehran University of Medical Sciences, Tehran,<br />

Iran.<br />

Received 20 May 2011; Accepted 07 March 2012<br />

Abstract<br />

Background: Diabetes mellitus is capable of impairing the myocardial function. Several studies have documented the<br />

influential impact of diabetes mellitus on the left ventricular function. The right ventricular function plays a significant role<br />

in the overall myocardial contractility; hence, this study was undertaken to evaluate the effect of diabetes mellitus type II on<br />

the right ventricular function.<br />

Methods: Twenty-two diabetic patients without any coronary artery disease, hypertension, or left ventricular dysfunction<br />

were studied. The right ventricular end diastolic diameter, tricuspid plane systolic excursion, right ventricular inflow Doppler<br />

parameters, longitudinal myocardial velocities, and deformation indices from the basal and apical segments of the right<br />

ventricular free wall of the case group were measured. The control group consisted of 22 healthy individuals.<br />

Results: The tricuspid annular plane systolic excursion (TAPSE) and tricuspid peak early to peak late diastolic flow<br />

velocities ratio (E/A) in the diabetic patients were significantly lower than those of the control group patients (18.9 vs. 23.2,<br />

p value < 0.001 and 0.96 vs. 1.21, p value = 0.012), but there were no significant differences in the right ventricular end<br />

diastolic diameter and the right ventricular Tei index between the two groups (p value = 0.72). The right ventricular basal<br />

peak myocardial systolic velocity (SM) (12 cm/sec vs. 13.4 cm/sec; p value = 0.03), basal and apical right ventricular free<br />

wall systolic strain (-13.3% and -18.7% vs. -20.2% and -25.7%; p value = 0.001), and apical strain rate (-1.2 1/s vs. -1.6 1/s;<br />

p value = 0.008 ) were significantly lower in the study group. There was a weak correlation between the right ventricular<br />

function and HbA1c as well as the duration of diabetes mellitus and C-reactive protein.<br />

Conclusion: Our results suggest that diabetes mellitus type II can influence the right ventricular function in the absence of<br />

coronary artery disease, diastolic dysfunction, and pulmonary hypertension.<br />

J Teh Univ Heart Ctr 2012;7(4):177-181<br />

This paper should be cited as: Parsaee M, Bahmanziari P, Ardeshiri M, Esmaeilzadeh M. Obvious or Subclinical Right Ventricular<br />

Dysfunction in Diabetes Mellitus (Type II): An Echocardiographic Tissue Deformation Study. J Teh Univ Heart Ctr 2012;7(4):177-181.<br />

Keywords: Echocardiography • Diabetes mellitus • Ventricular function, right<br />

*<br />

Corresponding Author: Mozhgan Parsaee, Assistant Professor of Cardiology, Department of Echocardiography, Rajaei Cardiovascular, Medical<br />

and Research Center, Vali-Asr Ave. Adjacent to Mellat Park, Tehran, Iran. 1996911151. Tel: +98 21 23922930. Fax: +98 21 2055594. E-mail:<br />

parsaeemozhgan@yahoo.com.<br />

The Journal of Tehran University Heart Center 177


The Journal of Tehran University Heart Center<br />

Introduction<br />

Diabetes mellitus (DM) increases the risk of heart failure<br />

development even in the absence of coronary artery disease,<br />

hypertension, or other comorbidities 1, 2 and could result in<br />

a two to fourfold greater mortality in heart failure patients.<br />

Indeed, DM is capable of impairing the myocardial function:<br />

this is initially a clinically silent condition; nevertheless, if<br />

left unrecognized and insufficiently managed, it could beget<br />

overt diabetic cardiomyopathy.<br />

The right ventricular (RV) function plays a significant<br />

role in the overall myocardial contractility. 3 Nevertheless,<br />

most of the previous studies regarding diabetes-induced<br />

changes in myocardial dysfunction were dedicated to the left<br />

ventricle (LV) at the cost of ignoring the role of the right<br />

heart chambers. The existing literature of course does contain<br />

a limited number of studies on the RV cardiomyopathies of<br />

patients with DM type I; however, to our knowledge, there<br />

is only one recent study probing into DM type II drawing up<br />

on three-dimensional strain and strain rate.<br />

The objectives of the present study were to evaluate the<br />

RV systolic and diastolic functions using conventional<br />

echocardiography, tissue Doppler imaging (TDI), and<br />

deformation indices (strain and strain rate) in patients with<br />

DM type II and without coronary artery disease or LV<br />

dysfunction.<br />

Methods<br />

The study group was comprised of 22 diabetic patients (9<br />

men and 13 women) aged 55.6 ± 6.0 years. These patients<br />

either referred to the DM Clinic of Rajaei Cardiovascular,<br />

Medical and Research Center or were amongst the hospital<br />

stuff. The control group consisted of 22 healthy individuals<br />

(9 men and 13 women). All the diabetic patients enrolled<br />

in this study were asymptomatic, without any clinical<br />

evidence of either systolic or diastolic heart failure. The<br />

control subjects were considered to have a low probability<br />

of coronary disease or heart failure based on clinical data.<br />

Coronary artery disease was excluded if there was a<br />

negative Dobutamine stress echocardiographic examination<br />

or a negative treadmill exercise electrocardiographic test.<br />

In a few cases, coronary artery disease was excluded on the<br />

evidence of equivocal non-invasive stress tests and normal<br />

coronary angiograms. The other exclusion criteria included<br />

valvular or congenital heart disease, left ventricular ejection<br />

fraction (LVEF) less than 50%, rhythms other than normal<br />

sinus rhythm, documented pulmonary diseases or pulmonary<br />

hypertension, and tricuspid regurgitation more than a mild<br />

degree.<br />

All the diabetic patients were on a diet and treatment with<br />

oral hypoglycemic drugs. Plasma glucose and HbA1c were<br />

observed periodically and controlled by the endocrinologist<br />

Mozhgan Parsaee et al.<br />

in the aforementioned clinic. None of the patients had such<br />

diabetic complications as progressive and complex diabetic<br />

retinopathy, neuropathy, and nephropathy.<br />

The study participants in the two groups underwent<br />

echocardiographic examinations (Vivid 7, GE Vingmed).<br />

The variables measured are presented as follows:<br />

1. The RV dimension and tricuspid annular plane systolic<br />

excursion (TAPSE) were measured from the apical fourchamber<br />

view.<br />

2. The RV Doppler parameters of the diastolic function<br />

(peak early [A] and peak late diastolic flow velocity [E],<br />

together with their ratio [E/A] and deceleration time [DT])<br />

were recorded. The trans-tricuspid flow was measured in the<br />

apical four-chamber view using pulsed Doppler, with the<br />

sample volume positioned at the tips of the tricuspid leaflets.<br />

The measurements were averaged from three end-expiratory<br />

cycles.<br />

3. TDI was conducted to assess the RV longitudinal<br />

myocardial function in the apical four-chamber view. Color<br />

TDI was used in addition to two-dimensional images,<br />

with adjustment of the depth and sector angle to obtain an<br />

acceptable frame rate. The TDI variables of peak systolic<br />

velocity (Sm), peak early diastolic velocity (Em), peak late<br />

diastolic velocity (Am), myocardial performance index<br />

(MPI), and acceleration time of the isovolumetric contraction<br />

time (IVCT) were analyzed online.<br />

4. The deformation indices were measured by adjusting the<br />

imaging angle to achieve a parallel alignment of the beam<br />

with the myocardial segment of interest. The images were<br />

stored on a remote archive of the echocardiography machine<br />

so that they could be analyzed offline. The myocardial<br />

deformation curves were investigated from the basal segment<br />

of the RV free wall, which belongs to the inflow part of the<br />

RV, and also from the apical segment, which belongs to the<br />

trabecular portion of the RV.<br />

5. The strain rate and stain were measured based on the<br />

standard deformation measurement, and the peak of the R<br />

on the electrocardiography (ECG) was utilized to define<br />

end diastole. End systole was determined using the pulsed<br />

Doppler velocity profile of the left ventricular outflow tract<br />

(LVOT) as the aortic valve closure, and the RV end systole<br />

was determined using the pulsed wave profile of the right<br />

ventricular outflow tract (RVOT) at end expiration. The<br />

analysis of the myocardial strain and strain rate data included<br />

the assessment of the systolic strain (є), peak systolic strain<br />

rate (SRs), and peak early diastolic strain rate (SRe).<br />

The data are expressed as Mean ± SD and percentages. The<br />

Student unpaired t-test was used to evaluate the differences<br />

between the groups, and the chi-square test was employed to<br />

compare the categorical variables. The Pearson correlation<br />

coefficients were utilized to pair the continuous variables.<br />

A two-tailed p value < 0.05 was considered significant. For<br />

the statistical analyses, the statistical software SPSS version<br />

17.0 for Windows (SPSS Inc., Chicago, IL) was used.<br />

178


Table 1. Demographic characteristics of the studied groups * Diabetic group Control subjects P value<br />

Table 2. Echocardiographic and Doppler data of the studied groups * Diabetic group Control subjects P value<br />

Obvious or Subclinical Right Ventricular Dysfunction in Diabetes Mellitus (Type II): ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Results<br />

The demographic characteristics and clinical parameters<br />

of the participants are depicted in Tables 1. There were no<br />

significant differences between the two groups in terms<br />

of sex, age, body mass index, creatinine, total cholesterol<br />

level, low-density lipoprotein cholesterol (LDL), highdensity<br />

lipoprotein cholesterol (HDL), and being a smoker.<br />

The diabetic group, however, showed a higher level of<br />

triglyceride (p value = 0.049), but this was statistically, and<br />

not clinically, significant.<br />

The echocardiographic and Doppler data of the studied<br />

groups are illustrated in Table 2. There were no significant<br />

differences between the two groups as regards the right<br />

ventricular end diastolic diameter (RVEDD) (mm), and<br />

tricuspid E velocity, whereas the tricuspid annular plane<br />

systolic excursion (TAPSE), and tricuspid E/A ratio of the<br />

diabetic patients (18.9 ± 2.1 vs. 23.2 ± 2.9 and 0.96 ± 0.2<br />

vs. 1.21 ± 0.3, respectively) were significantly lower. The<br />

tricuspid A velocity and E wave deceleration time of the<br />

diabetic patients (0.5 ± 0.1 m/s vs. 0.4 ± 0.1 m/s and 289.2<br />

± 67 msec vs. 250.9 ± 57.1 msec) were significantly higher.<br />

Age (y) 54.5±7.0 49.8±8.4 0.05<br />

Male 9 (40.9) 9 (40.9) 0.69<br />

Hypertension 9 (40.9) 4 (18.2) 0.07<br />

Smoking 2 (9.1) 3 (13.6) 0.82<br />

Body mass index (Kg/m 2 ) 28.7±3.1 26.4±3.8 0.05<br />

Systolic blood pressure (mm Hg) 125.6±15.4 120.5±13.4 0.28<br />

Diastolic blood pressure (mm Hg) 79.5±11.6 76.1±10.5 0.32<br />

Creatinine (mg/dl) 1.0±0.2 1.1±0.1 0.47<br />

Total cholesterol (mg/dl) 178.4±40.0 166.0 ±37.0 0.39<br />

Duration of diabetes (y) 7.0±3.0 - -<br />

Triglyceride (mg/dl) 186.0±82.0 130.0±67.0 0.04<br />

Low-density lipoprotein cholesterol (mg/dl) 90.0±26.0 89.0±27.0 0.96<br />

*<br />

Data are presented as mean±SD or n (%)<br />

Right ventricular end diastolic diameter (mm) 28.0±2.9 28.0±3.7 0.56<br />

Tricuspid annular plane systolic excursion (mm) 18.9±2.1 23.2±2.9 < 0.01<br />

Tricuspid E velocity (m/s) 0.47±0.05 0.46±0.08 0.52<br />

Tricuspid A velocity (m/s) 0.5±0.1 0.4±0.1 0.04<br />

Tricuspid deceleration time (msec) 289.2±67.0 250.9±57.1 0.04<br />

E/A ratio 0.9±0.2 1.2±0.3 0.01<br />

*<br />

Data are presented as mean±SD<br />

E/A, Tricuspid peak early to peak late diastolic flow velocities ratio<br />

Table 3. Tissue Doppler imaging data of the studied groups * Diabetic group Control subjects P value<br />

Right ventricular basal segment Sm (cm/s) 12.0±1.9 13.4±2.1 0.03<br />

Right ventricular basal segment Em (cm/s) 8.5±2.1 11.6±2.4 < 0.01<br />

Right ventricular basal segment Am (cm/s) 12.6±2.7 14.7±3.3 0.03<br />

Myocardial performance index 0.42±0.11 0.40±0.09 0.72<br />

Acceleration time of the isovolumetric contraction time (ms) 37.8±8.4 35.4±8.4 0.45<br />

E/Em ratio 0.05±0.01 0.04±0.01 < 0.01<br />

*<br />

Data are presented as mean±SD<br />

Sm, Peak myocardial systolic velocity; Em, Peak early diastolic velocity; Am, Peak late diastolic velocity; E, Peak late diastolic flow velocity<br />

The Journal of Tehran University Heart Center179


The Journal of Tehran University Heart Center<br />

Mozhgan Parsaee et al.<br />

Table 4. Deformation indices data of the studied groups * Diabetic group Control subjects P value<br />

Right ventricular basal Є (%)<br />

Right ventricular basal SRs (1/s)<br />

Right ventricular basal SRe (1/s)<br />

Right ventricular apical Є (%)<br />

Right ventricular apical SRs (1/s)<br />

Right ventricular apical SRe (1/s)<br />

*<br />

Data are presented as mean±SD<br />

Є, Systolic stain; SRs, Peak systolic strain rate; SRe, Early diastolic strain rate<br />

-13.3±4.3 -20.2±7.3 < 0.01<br />

-1.0±0.5 -1.37±0.9 0.18<br />

0.9±0.3 1.2±0.5 0.05<br />

-18.7±4.3 -25.7±8.2 < 0.01<br />

-1.2±0.3 -1.6±0.6 < 0.01<br />

1.2±0.4 1.9±0.7 < 0.01<br />

The TDI data of the study population are shown in Table<br />

3, according to which the RV basal segment Sm, Em, Am,<br />

and E/Em (12 ± 1.9 cm/sec vs. 13.4 ± 2.1 cm/sec, 8.5 ± 2.1<br />

cm/sec vs. 11.6 ± 2.4 cm/sec, 12.6 ± 2.7 cm/sec vs. 14.7 ±<br />

3.3 cm/sec, and 0.05 ± 0.01 vs. 0.04 ± 0.01, respectively)<br />

were significantly lower in the case group than in the control<br />

group. There were no significant differences between the two<br />

groups with respect to the MPI of the RV and the acceleration<br />

time of the IVCT.<br />

The data on the deformation indices of the two study<br />

groups are demonstrated in Table 4, according to which the<br />

RV basal segment systolic strain (Є) and RV apical segment<br />

(Є, SRs, and SRe) of the case group were significantly lower<br />

than those in the control group (p value < 0.01). There were,<br />

however, no significant differences in regard to the RV basal<br />

segment SRs and SRe between the two groups.<br />

There were no significant correlations between HBA1c,<br />

duration of diabetes, C-reactive protein (CRP), and measures<br />

of the RV systolic or diastolic dysfunction (based on<br />

conventional Doppler, TDI data, or deformation indices).<br />

Discussion<br />

The present study shows that both systolic and diastolic<br />

RV functions are impaired in patients with DM type II and<br />

without coronary artery disease or ejection fractions of less<br />

than 50%. This finding was demonstrated by significantly<br />

lower RV systolic parameters (TAPSE, RV basal Sm, basal<br />

and apical RV free wall systolic strains, and apical SRs) and<br />

lower RV diastolic parameters (E/A ratios, Em, Am, E/Em<br />

ratios, and apical early diastolic strain rate [SRe]) in our case<br />

group than those in a normal, sex- and age-matched control<br />

group. There were no statistically significant differences in<br />

the RV basal segment SRs and SRe between the diabetic and<br />

control subjects, which may be due to difficulties in drawing<br />

basal RV strain rate curves.<br />

Our results chime in with those of the Kosmala et al., 4<br />

study, which reported the impairment of both basal and apical<br />

segments of the RV free wall performance. In that study,<br />

however, the apical segments exhibited more pronounced<br />

systolic impairment than did the basal segments and it<br />

was concluded that the RV might be divided into the three<br />

components of the inflow or sinus, the trabecular, and the<br />

outflow portions. Geva et al., 5 stated that the inflow region,<br />

compared with the other parts of the RV, had significant<br />

predominance in fiber shortening and contribution to the<br />

global RV systolic function. As the basal segment of the RV<br />

free wall is a part of the inflow component, and the apical<br />

segment refers to the trabecular portion of the RV, they<br />

suggested that the differences seen in their cohort might be<br />

related to the regional inhomogeneity of the RV in patients<br />

with DM.<br />

The Kosmala et al., study 4 reported RV diastolic<br />

dysfunction in a non-uniform type I and type II diabetic<br />

cohort using TDI and demonstrated significantly lower<br />

values of Em and Em-to-Am ratio in the basal and midsegments<br />

and longer isovolumic relaxation time (IRTm) in<br />

the mid-segment. Nonetheless, that study failed to show<br />

any difference in the E/A ratio, Em, and systolic parameters<br />

(Sm and TAPSE) between the diabetic patients and normal<br />

subjects. Furthermore, the said study found no significant<br />

correlations between the estimated echocardiographic<br />

parameters and indices of diabetic control (plasma glucose<br />

and HbA1c) as well as the duration of diabetes.<br />

In another study, Kosmala et al., 4 evaluated non-uniform<br />

type I and type II diabetic groups and showed impairment<br />

of the RV systolic function according to deformation studies<br />

(RV systolic stain and SRs in the basal and apical segments<br />

of the RV free wall) and impairment of the RV diastolic<br />

function (decreased SRe in both RV segments). In a study by<br />

Karamitsos et al., 6 type I diabetic patients were found to have<br />

an impaired RV diastolic function (trans-tricuspid E velocity,<br />

A velocity, E/A ratio, Em, and Am). Nevertheless, the RV<br />

systolic function was preserved in the authors’ diabetic<br />

population.<br />

Consistent with the findings of the present study, Gaber<br />

et al., 7 assessed patients with DM type II and demonstrated<br />

impairment of the RV systolic and diastolic functions<br />

according to deformation studies via three-dimensional<br />

echocardiography (systolic strain, SRs, and SRe in both RV<br />

basal and apical segments).<br />

The present study, in agreement with some other similar<br />

studies, 4, 8, 9 found no importance for the impact of the<br />

180


Obvious or Subclinical Right Ventricular Dysfunction in Diabetes Mellitus (Type II): ...<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

duration of diabetes on the RV function. This finding could<br />

suggest that controlling diabetes mellitus is more important<br />

than its duration.<br />

Med J 2008;11:65-74.<br />

9. Rowland TW, Martha PM, Jr, Reiter EO, Cunningham LN. The<br />

influence diabetes mellitus on cardiovascular function in children<br />

and adolescents. Int J Sports Med 1992;13:431-435.<br />

Conclusion<br />

Whether the RV diastolic abnormalities have prognostic<br />

implications in the clinical course of patients with DM<br />

type II remains to be investigated. We believe that serial<br />

echocardiography measurements are warranted in this<br />

diabetic population if the progression from subclinical<br />

RV involvement to symptomatic RV dysfunction is to be<br />

followed. In addition, subclinical RV systolic and diastolic<br />

abnormalities should be considered when planning<br />

pharmacotherapy to prevent the development of symptomatic<br />

RV dysfunction.<br />

Acknowledgments<br />

This research project was supported by Rajaei<br />

Cardiovascular, Medical and Research Center. Special<br />

thanks are due to the above-mentioned hospital’s personnel<br />

and nurses who enrolled in our control group.<br />

References<br />

1. Kosmala W, Colonna P, Przewlocka-Kosmala M, Mazurek W.<br />

Right ventricular dysfunction in asymptomatic diabetic patients. J<br />

Diabetes Care 2004;27:2736-2738.<br />

2. Dounis V, Siegmund T, Hansen A, Jensen J, Schumm-Draeger PM,<br />

von Bibra H. Global myocardial perfusion and diastolic function<br />

are impaired to a similar extent in patients with type 2 diabetes<br />

mellitus and in patients with coronary artery disease-evaluation by<br />

contrast echocardiography and pulsed tissue Doppler. Diabetologia<br />

2006;49:2729-2740.<br />

3. Van den Brom CE, Bosmans JW, Vlasblom R, Handoko LM,<br />

Huisman MC, Lubberink M, Molthoff CF, Lammertsma AA,<br />

Ouwens MD, Diamant M, Boer C. Diabetic cardiomyopathy in<br />

Zucker diabetic fatty rats: the forgotten right ventricle. J Cardiovasc<br />

Diabetologia 2010;9:25-32.<br />

4. Kosmala W, Przewlocka-Kosmala M, Mazurek W. Subclinical<br />

right ventricular dysfunction in diabetes mellitus-an ultrasonic<br />

strain/strain rate study. Diabet Med 2007;24:656-663.<br />

5. Geva T, Powell AJ, Crawford EC, Chung T, Colan SD. Evaluation<br />

of regional differences in right ventricular systolic function by<br />

acoustic quantification by echocardiography and cine magnetic<br />

resonance imaging. Circulation 1998;98:339-345.<br />

6. Karamitsos TD, Karvounis HI, Dalamanga EG, Papadopoulos<br />

CE, Didangellos TP, Karamitsos DT, Parharidis GE, Louridas GE.<br />

Early diastolic impairment of diabetic heart: the significance of<br />

right ventricle. Int J Cardiol 2007;114:218-223.<br />

7. Gaber R, Kotb NA. Early diagnosis of right ventricular dysfunction<br />

in type II diabetes mellitus: value of 3 dimensional strain/strain<br />

rate. Heart mirror J 2010;4:80-85.<br />

8. Elshahed GS, Ahmed MI, El-Beblawy NS, Kamal HM, lsmaiel<br />

MF, Bin Zheidan OAS. Evaluation of right and left ventricular<br />

systolic and diastolic function in patients with type I diabetes using<br />

echocardiography and tissue Doppler imaging. Suez Canal Univ<br />

The Journal of Tehran University Heart Center181


The Journal of Tehran University Heart Center<br />

Case Report<br />

Surgical Treatment of Amplatzer Embolus in a Secundum<br />

Atrial Septal Defect Patient<br />

Ahmet Baris Durukan, MD 1* , Hasan Alper Gurbuz, MD 1 , Murat Tavlasoglu,<br />

MD 2 , Nevriye Salman, MD 1 , Halil Ibrahim Ucar, MD 1 , Cem Yorgancioglu,<br />

MD 1<br />

1<br />

Medicana International Ankara Hospital, Ankara, Turkey.<br />

2<br />

Diyarbakir Military Hospital, Diyarbakir, Turkey.<br />

Received 18 August 2011; Accepted 23 January 2012<br />

Abstract<br />

A secundum atrial septal defect is the most common congenital heart defect. Transcatheter treatment of secundum atrial<br />

septal defects is a popular and less invasive alternative to surgery. Procedural complications may occur in a wide spectrum,<br />

particularly device embolus as the most emergent one, but luckily they do not commonly occur in the clinical setting. Mortality<br />

from adverse events related to transcatheter treatment strategies is twentyfold higher than that of primary elective surgical<br />

closure. Here, we report an Amplatzer device embolus in a secundum atrial septal defect patient. The device was successfully<br />

removed with surgery, postoperative course was uneventful, and the patient was discharged from the hospital on the 5 th<br />

postoperative day.<br />

J Teh Univ Heart Ctr 2012;7(4):182-184<br />

This paper should be cited as: Durukan AB, Gurbuz HA, Tavlasoglu M, Salman N, Ucar HI, Yorgancioglu C. Surgical Treatment of<br />

Amplatzer Embolus in a Secundum Atrial Septal Defect Patient. J Teh Univ Heart Ctr 2012;7(4):182-184.<br />

Keywords: Heart septal defects, atrial • Septal occluder device • Surgical procedures, operative<br />

Introduction<br />

A secundum atrial septal defect (ASD) is the most<br />

common congenital heart defect, making up 10% of all<br />

congenital heart defects. Five to ten percent of children and<br />

30% of adults with congenital heart disease have the ostium<br />

secundum ASD. 1 ASD closure is indicated to prevent right<br />

ventricular volume overload due to left-to-right shunting<br />

over four years of age and to prevent arrhythmias and right<br />

heart failure in later decades. 2<br />

Transcatheter treatment of the secundum ASD is a popular<br />

and less invasive alternative to surgery. By 2008, 30000<br />

devices had been implanted. 2 Procedural complications<br />

do exist, luckily very rarely in the clinical setting. These<br />

complications increase morbidity and mortality rates<br />

compared to conventional surgical closure. 3<br />

Here, we report a case with the secundum ASD in which<br />

the atrial septal occluder device was embolized during the<br />

transcatheter procedure and was successfully treated with<br />

surgery.<br />

Case Report<br />

A 38-year-old, 160 cm tall, 65 kg woman was referred to<br />

our hospital with the diagnosis of the secundum ASD. She<br />

*<br />

Corresponding Author: Ahmet Baris Durukan, Medicana International Ankara Hospital, Umit Mahallesi 2463.sokak 4/18, 06810, Yenimahalle,<br />

Ankara, Turkey. Tel: +90 532 2273814. Fax: +90 312 2203170. E-mail: barisdurukan@yahoo.com.<br />

182


Surgical Treatment of Amplatzer Embolus in a Secundum Atrial Septal Defect Patient<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

had initially referred to another hospital with the complaints<br />

of palpitation and shortness of breath. During cardiologic<br />

evaluation, echocardiography was performed and a secundum<br />

type ASD (pulmonary to systemic blood flow ratio [Qp/Qs]:<br />

2.0), first to second degree mitral regurgitation, and first<br />

to second degree tricuspid regurgitation were noted. There<br />

was a mild increase in the right ventricular dimensions.<br />

Pulmonary artery pressure was measured as 45 mm Hg. In<br />

our hospital, echocardiography was performed again and<br />

the anatomy was found suitable for the implantation of an<br />

ASD occluder device. During catheter closure, the occluder<br />

device was embolized. The patient was heparinized for<br />

systemic anticoagulation. It was not possible to retrieve<br />

the embolized device via catheterization procedures, and<br />

the patient was referred for emergency surgical removal.<br />

Surgical intervention was performed within forty-five<br />

minutes of embolization. The electrocardiogram was normal<br />

sinus rhythm. On chest X-ray, the ‘Amplatzer occluder<br />

Device’ was seen in the right pulmonary artery localization<br />

(Figure 1).<br />

was closed with a Teflon patch. Right pulmonary arteriotomy<br />

was performed proximal to the superior vena cava and the<br />

device was removed (Figures 2 and 3). Arteriotomy was<br />

closed primarily. The postoperative course was uneventful,<br />

and the patient was discharged on the 5 th postoperative day.<br />

Figure 2. Intraoperative image. The right pulmonary artery is opened and<br />

the septal occluder device (arrow) is being removed through the arteriotomy<br />

Figure 3. Removed "Amplatzer Occluder Device"<br />

Figure 1. Preoperative chest X-ray of the patient, performed following device<br />

embolization. The arrow points very clearly to the septal occluder device<br />

in the right pulmonary artery<br />

The patient underwent surgery under general anesthesia<br />

and with standard median sternotomy incision. There were<br />

pericardial adhesions. Once the pericardium had been<br />

opened, the ‘Amplatzer Occluder Device’ was easily palpated<br />

in the right pulmonary artery proximal to the superior vena<br />

cava. Cardiopulmonary bypass (CPB) under moderate<br />

hypothermia was established with aorto-bicaval cannulation<br />

(selective superior vena cava cannulation was performed).<br />

Myocardial management was provided by antegrade cold<br />

and terminal warm blood cardioplegia. Right atriotomy was<br />

performed. A 2.5 × 1.5 cm secundum type ASD was seen. It<br />

Discussion<br />

The approach has shifted from surgical closure to<br />

transcatheter procedures in the treatment of the secundum<br />

ASD, which is definitely a less invasive approach. Hospital<br />

stay is shorter with transcatheter treatment and cosmetic<br />

results are better. Return to work for adults and return to<br />

school for children are earlier than those with cardiac surgery.<br />

Developmental outcome has been reported to be better in<br />

cases treated with transcatheter procedures. Adverse events<br />

related with CPB are avoided. 4 Right ventricular functions<br />

are preserved better. 5 And most importantly, the incidence<br />

of minor or major complications is lower with transcatheter<br />

closure. 6<br />

Procedural complications like device malposition or<br />

embolization, atrial wall erosion, mitral regurgitation,<br />

The Journal of Tehran University Heart Center183


The Journal of Tehran University Heart Center<br />

Ahmet Baris Durukan et al.<br />

cardiac arrhythmia requiring treatment, sciatic nerve<br />

compression due to retroperitoneal hematoma, prolonged<br />

time for femoral vein hemostasis, access site hematoma,<br />

arteriovenous fistula formation, pericardial/pleural<br />

effusions, and transient ischemic attacks have been<br />

previously reported. 1, 2 Butera et al. 1 reported 0.7%<br />

malposition/embolization. Wilson et al. 2 reported device<br />

embolization in 2 out of 227 patients and one was again<br />

treated via the transcatheter route. Sarris et al. 7 reviewed<br />

data from 19 well-known European institutes; and in a 10-<br />

year period, 56 cases undergoing emergency surgery after<br />

catheter intervention were reported. Among the 56 cases,<br />

there were 22 embolization cases and 7 cases were due<br />

to the pulmonary artery. Surgical removal of the device,<br />

closure of the defect, and repair/replacement of the damaged<br />

cardiac structure were performed. Operative mortality was<br />

5.4%. Based on the EACTS reports, operative mortality<br />

for primary surgical ASD closure was 0.36% in the same<br />

time period. 7 Based on the FDA reports, in a 7-year period,<br />

mortality rates for transcatheter closure were similar to<br />

those of primary surgical closure (0.13%), but the mortality<br />

for the surgical management of a device adverse event<br />

(2.6%) was twentyfold higher than that for primary elective<br />

ASD closure. 3<br />

We managed to remove the device in a very short time, and<br />

the patient was discharged without any further complications.<br />

of the US Food and Drug Administration Manufacturer and User<br />

Facility Device Experience database for adverse events involving<br />

Amplatzer septal occluder devices and comparison with the<br />

Society of Thoracic Surgery congenital cardiac surgery database.<br />

J Thorac Cardiovasc Surg 2009;137:1334-1341.<br />

4. Visconti KJ, Bichell DP, Jonas RA, Newburger JW, Bellinger<br />

DC. Developmental outcome after surgical versus interventional<br />

closure of secundum atrial septal defect in children. Circulation<br />

1999;100:II145-150.<br />

5. Dhillon R, Josen M, Henein M, Redington A. Transcatheter closure<br />

of atrial septal defect preserves right ventricular function. Heart<br />

2002;87:461-465.<br />

6. Berger F, Vogel M, Alexi-Meskishvili V, Lange PE. Comparison of<br />

results and complications of surgical and Amplatzer device closure<br />

of atrial septal defects. J Thorac Cardiovasc Surg 1999;118:674-<br />

678.<br />

7. Sarris G, Kirvassilis G, Zavaropoulos P, Belli E, Berggren H, Carrel<br />

T, Comas JV, Corno AF, Daenen W, Di Carlo D, Ebels T, Fragat J,<br />

Hamilton L, Hraska V, Jacobs J, Lazarov S, Mavroudis C, Metras<br />

D, Rubay J, Schreiber C, Stellin G. Surgery for complications of<br />

transcatheter closure of atrial septal defects: a multi-institutional<br />

study from the European Congenital Heart Surgeons Association.<br />

Eur J Cardiothorac Surg 2010;37:1285-1290.<br />

Conclusion<br />

Transcatheter device closure of the ASD can lead to serious<br />

complications despite its advantages over surgical closure.<br />

But it should be kept in mind that even if most cases are<br />

successful, not only early, but also late complications may<br />

occur regardless of the size or type of the current devices.<br />

Surgical treatment of the complications may be mandatory<br />

and should be performed immediately, especially in cases<br />

of embolization. Surgery is quite effective in treatment,<br />

but it is a fact that operative mortality rises when surgery<br />

is performed for the treatment of complications. Operative<br />

mortality is much lower for primary surgical repair.<br />

References<br />

1. Butera G, Romagnoli E, Carminati M, Chessa M, Piazza L, Negura<br />

D, Giamberti A, Abella R, Pome G, Condoluci C, Frigiola A.<br />

Treatment of isolated secundum atrial septal defects: impact of age<br />

and defect morphology in 1,013 consecutive patients. Am Heart J<br />

2008;156:706-712.<br />

2. Wilson N, Smith J, Prommete B, O’Donnell C, Gentles TL,<br />

Ruygrok PN. Transcatheter closure of secundum atrial septal<br />

defects with the Amplatzer septal occluder in adults and childrenfollow-up<br />

closure rates, degree of mitral regurgitation and<br />

evolution of arrhythmias. Heart Lung Circ 2008;17:318-324.<br />

3. DiBardino DJ, McElhinney DB, Kaza AK, Mayer JE, Jr. Analysis<br />

184


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Case Report<br />

Post-Traumatic Chordae Rupture of Tricuspid Valve<br />

Kyomars Abbasi, MD, Hossein Ahmadi, MD, Arezoo Zoroufian, MD,<br />

Mohammad Sahebjam, MD, Naghmeh Moshtaghi, MD, Seyed Hessamedin<br />

Abbasi, MD *<br />

Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.<br />

Received 26 October 2011; Accepted 07 January 2012<br />

Abstract<br />

Blunt injury to the chest can affect any one or all components of the chest wall and thoracic cavity. The clinical presentation<br />

of patients with blunt chest trauma varies widely and ranges from minor reports of pain to florid shock. Traumatic tricuspid<br />

valve regurgitation is a rare cardiovascular complication of blunt chest trauma. Tricuspid valve regurgitation is usually<br />

begotten by disorders that cause the right ventricle to enlarge. Diagnosis is made by physical examination findings and is<br />

confirmed by echocardiography. We report two cases of severe tricuspid regurgitation secondary to the rupture of the chordae<br />

tendineae of the anterior leaflet following non-penetrating chest trauma. Both patients had uneventful postoperative courses.<br />

J Teh Univ Heart Ctr 2012;7(4):185-187<br />

This paper should be cited as: Abbasi K, Ahmadi H, Zoroufian A, Sahebjam M, Moshtaghi N, Abbasi SH. Post-Traumatic Chordae<br />

Rupture of Tricuspid Valve. J Teh Univ Heart Ctr 2012;7(4):185-187.<br />

Keywords: Heart rapture • Tricuspid valve • Wounds and injuries<br />

Introduction<br />

Blunt thoracic trauma has become a regular occurrence<br />

courtesy of high-speed motor vehicle accidents, 1 especially<br />

in the last decade. 2 Such accidents are the principal cause of<br />

isolated tricuspid valve regurgitation (TR). 3 Traumatic TR<br />

is a rare cardiovascular complication of blunt chest trauma.<br />

The most frequently involved valve in blunt chest trauma is<br />

the aortic valve, followed by the mitral and tricuspid valves. 4<br />

Traumatic TR is usually well tolerated; 5 however, as far as<br />

treatment is concerned, the tricuspid valve replacement has<br />

been the conventional procedure, especially in cases with<br />

delayed presentation. 6 We herein report two cases of severe<br />

TR secondary to the rupture of the chordae tendineae of the<br />

anterior leaflet following non-penetrating chest trauma.<br />

Case Reports<br />

Case # 1<br />

A 28-year-old man was admitted to our hospital suffering<br />

from exertional dyspnea and palpitation. One year previously,<br />

he had sustained blunt chest trauma in a car accident, but<br />

no cardiac abnormality had been noticed at the time. His<br />

symptoms started about two months before his referral to us.<br />

At presentation, the patient had a blood pressure of 120/90<br />

mmHg and pulse of 70 beats/min. The electrocardiogram<br />

revealed sinus rhythm and right bundle branch block with<br />

left-axis deviation. Transthoracic echocardiography (TTE)<br />

demonstrated a normal left ventricle (LV) size, mild LV<br />

systolic dysfunction, septal motion abnormality due to the<br />

right ventricle (RV) volume overload, severe RV and right<br />

*<br />

Corresponding Author: Seyed-Hesameddin Abbasi, Tehran University of Medical Sciences, Tehran Heart Center, North Kargar Street, Tehran, Iran.<br />

1411713138. Tel: +98 21 88029720. Fax: +98 21 88029702. E-mail: abbasi@tehranheartcenter.org.<br />

The Journal of Tehran University Heart Center 185


The Journal of Tehran University Heart Center<br />

atrium (RA) enlargement, and finally, severe low pressure<br />

TR due to the flail anterior leaflet of the tricuspid valve.<br />

Transesophageal echocardiography (TEE) was conducted<br />

as a complementary study and confirmed the flail leaflet<br />

secondary to the ruptured chordae. The patient was, therefore,<br />

scheduled for tricuspid valve repair.<br />

The operation was performed through median sternotomy.<br />

Following the institution of cardiopulmonary bypass (CPB),<br />

the heart was arrested via cardioplegia and the RA was<br />

explored: there was chordae rupture of the anterior leaflet.<br />

The rupture was repaired by first constructing two near<br />

chordae with 5/0 Gortex suture and then placing edge-toedge<br />

stitches on the edge of the three leaflets. Additionally,<br />

ring annuloplasty using a Carpenter (31mm) was performed.<br />

Postoperative TEE demonstrated mild to moderate TR and<br />

no significant tricuspid valve stenosis (TS) (tricuspid valve<br />

mean gradient = 2.5 mm Hg). The patient’s postoperative<br />

course was uneventful.<br />

Case # 2<br />

A 53-year-old man was referred to our hospital because of<br />

exertional dyspnea. One year previously, he had sustained<br />

multiple traumas in a bus accident. The accident left him in<br />

a deep coma for 17 days, after which he suffered a stroke,<br />

right hemi-paresis, and aphasia due to infarction in the left<br />

temporal lobe. Also, he had fractures in the left femoral<br />

bone, left tibia bone, right hand, and two ribs on the superior<br />

right side. At the time, no cardiac symptoms were detected<br />

but the patient was subjected to splenectomy.<br />

On admission, the electrocardiogram showed sinus rhythm,<br />

right bundle branch block with left-axis deviation. TTE<br />

revealed a normal LV size with mildly reduced LV systolic<br />

function, abnormal septal motion due to the RV volume<br />

overload, severe RV and RA enlargement with mild RV<br />

systolic dysfunction, and flail anterior tricuspid valve leaflet<br />

with severe low pressure TR. In addition, a large, mobile<br />

filamentous mass was detected on the tip of the flail leaflet, in<br />

favor of ruptured chordae. These findings were all confirmed<br />

by preoperative TEE, which also demonstrated an aneurismal<br />

inter-atrial septum with a small secondary type of atrial septal<br />

defect (ASD). Cardiac catheterization showed a small left-toright<br />

shunt with no evidence of coronary artery disease.<br />

Intraoperatively, the chest was explored via median<br />

sternotomy. CPB was instituted, the heart was arrested by<br />

cardioplegia, and the RA was explored. The anterior papillary<br />

muscle was ruptured, resulting in the prolapse of the anterior<br />

tricuspid leaflet. The De Vega technique was applied to the<br />

tricuspid annulus. Pledgeted 4/0 Prolene suture was used for<br />

the reconstruction of the anterior tricuspid valve papillary<br />

muscle to scar tissue in the base of the papillary muscle.<br />

Linear repair was thereafter performed for the closure of<br />

the ASD. Saline test was carried out next and the result was<br />

Kyomars Abbasi et al.<br />

so optimal that it obviated the need for TEE. Postoperative<br />

TTE revealed mild RV enlargement with mild systolic<br />

dysfunction. Doppler and contras study showed mild TR and<br />

no residual shunt flow.<br />

The patient made an uneventful recovery. One year after<br />

surgery now, the patient is well and asymptomatic with<br />

trivial TR on echocardiography.<br />

Discussion<br />

Traumatic TR is a rare sequela in the wake of blunt thoracic<br />

trauma. The most likely cause is compression, decompression,<br />

or deceleration of the thorax. These mechanisms give rise to<br />

intraventricular pressure peaks, which in combination with a<br />

closed valve and the systolic pressure in the RV most often<br />

affect the chordae tendineae and the papillary muscles. 1<br />

TR frequently goes undiagnosed at blunt chest trauma<br />

because of its asymptomatic nature; sometimes several<br />

or even more than ten years elapse before a diagnosis is<br />

made. 7-10 Nevertheless, severe TR eventually leads to right<br />

ventricular dilatation and progressive right heart failure,<br />

even if it is associated with no pulmonary hypertension. 6 The<br />

clinical course of TR following blunt chest trauma varies<br />

largely. Little is known about the clinical course of isolated<br />

ruptured chordae tendineae. Indeed, the only detailed case to<br />

have been reported thus far is that by Kleikamp et al., whose<br />

patient developed TR due to ruptured chordae tendineae of<br />

the anterior leaflet seven years after a blunt chest trauma,<br />

which was surgically corrected. 1 It is deserving of note<br />

that the rupture of the chordae tendineae is believed to be<br />

the consequence of an acute increase in the RV pressure<br />

associated with a closed tricuspid valve. 11<br />

Conclusions<br />

The two cases presented herein demonstrate that the<br />

rupture of the chordae tendineae of the tricuspid valve could<br />

be another cause of TR following blunt chest trauma. This<br />

is, however, a condition that can be repaired surgically<br />

without the need for valve replacement. Advances in<br />

echocardiography have enabled earlier diagnosis and ergo<br />

more effective treatment.<br />

We recommend that physicians working at emergency<br />

departments be on the alert for this potential complication<br />

of non-penetrating chest trauma and subject all patients<br />

admitted to the emergency department due to blunt chest<br />

trauma to TTE or TEE for accurate diagnosis.<br />

References<br />

1. Kleikamp G, Schnepper U, Körtke H, Breymann T, Körfer R.<br />

186


Post-Traumatic Chordae Rupture of Tricuspid Valve<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Tricuspid valve regurgitation following blunt thoracic trauma.<br />

Chest 1992;102:1294-1296.<br />

2. Lin SJ, Chen CW, Chou CJ, Liu KT, Su HM, Lin TH, Voon WC,<br />

Lai WT, Sheu SH. Traumatic tricuspid insufficiency with chordae<br />

tendinae rupture: a case report and literature review. Kaohsiung J<br />

Med Sci 2006;22:626-629.<br />

3. Nelson M, Wells G. A case of traumatic tricuspid valve<br />

regurgitation caused by blunt chest trauma. J Am Soc Echocardiogr<br />

2007;20:198.e4-5.<br />

4. Colombo T, Russo C, Ciliberto GR, Lanfranconi M, Bruschi G,<br />

Agati S, Vitali E. Tricuspid regurgitation secondary to mitral valve<br />

disease: tricuspid annulus function as guide to tricuspid valve<br />

repair. Cardiovasc Surg 2001;9:369-377.<br />

5. Noera G, Sanguinetti M, Pensa P, Biagi B, Cremonesi A, Lodi R,<br />

Lessana A, Carbone C. Tricuspid valve incompetence caused by<br />

nonpenetrating thoracic trauma. Ann Thorac Surg 1991;51:320-<br />

322.<br />

6. Fujiwara K, Hisaoka T, Komai H, Nishimura Y, Yamamoto<br />

Sh, Okamura Y. Successful repair of traumatic tricuspid valve<br />

regurgitation. Jpn J Thorac Cardiovasc Surg 2005;53:259-262.<br />

7. Gayet C, Pierre B, Delahaye JP, Champsaur G, Andre-Fouet X,<br />

Rueff P. Traumatic tricuspid insufficiency. An underdiagnosed<br />

disease. Chest 1987;92:429-432.<br />

8. van Son JA, Danielson GK, Schaff HV, Miller FA, Jr. Traumatic<br />

tricuspid valve insufficiency. Experience in thirteen patients. J<br />

Thorac Cardiovasc Surg 1994;108:893-898.<br />

9. Holper K, Hahnel C, Augustin N, Meisner H. Operative correction<br />

of traumatic tricuspid insufficiency. Herz 1996;21:172-178.<br />

10. Maisano F, Lorusso R, Sandrelli L, Torracca L, Coletti G, La Canna<br />

G, Alfieri O. Valve repair for traumatic tricuspid regurgitation. Eur<br />

J Cardiothorac Surg 1996;10:867-873.<br />

11. dos Santos J, Jr, de Marchi CH, Bestetti RB, Corbucci HA,<br />

Pavarino PR. Ruptured chordae tendineae of the posterior leaflet of<br />

the tricuspid valve as a cause of tricuspid regurgitation following<br />

blunt chest trauma. Cardiovasc Pathol 2001;10:97-98.<br />

The Journal of Tehran University Heart Center187


The Journal of Tehran University Heart Center<br />

Case Report<br />

Late Diagnosis of Large Left Ventricular Pseudoaneurysm<br />

after Mitral Valve Replacement and Coronary Artery<br />

Bypass Surgery by Real-Time Three-Dimensional<br />

Echocardiography<br />

Mohammad Sahebjam, MD * , Abbas Salehiomran, MD, Neda Ghaffari-Marandi,<br />

MD, Azam Safir, MD<br />

Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.<br />

Abstract<br />

One of the most serious complications of mitral valve replacement is left ventricular rupture and pseudoaneurysm<br />

formation, which is rare but potentially lethal. We herein present a late type of post mitral valve replacement and coronary<br />

artery bypass surgery pseudoaneurysm in a 74-year-old female, who was admitted to our hospital with a recent history of<br />

exertional dyspnea. She had the above-mentioned operation 10 months before. The diagnosis was made via two-dimensional<br />

and real-time three-dimensional transthoracic echocardiography. The prosthetic mitral valve was removed, and the large<br />

orifice of the pseudoaneurysm was closed by surgery. At one year's follow-up, the patient was in good condition.<br />

J Teh Univ Heart Ctr 2012;7(4):188-190<br />

Received 17 August 2011; Accepted 04 February 2012<br />

This paper should be cited as: Sahebjam M, Salehiomran A, Ghaffari-Marandi N, Safir A. Late diagnosis of Large Left Ventricular<br />

Pseudoaneurysm after Mitral valve Replacement and Coronary Artery Bypass Surgery by Real-Time Three-Dimensional Echocardiography.<br />

J Teh Univ Heart Ctr 2012;7(4):188-190.<br />

Keywords: Echocardiography, three-dimensional • Mitral valve • Aneurysm, false<br />

Introduction<br />

One of the most serious complications of mitral valve<br />

replacement (MVR) is the left ventricular (LV) rupture and<br />

pseudoaneurysm formation, which is rare but potentially<br />

lethal. 1-4 According to the time of the LV rupture, it is<br />

classified into three categories: early; delayed; and late<br />

ruptures. Early types are those that happen in the operating<br />

room after cardiopulmonary bypass discontinuation,<br />

delayed types are events which occur several hours or days<br />

after surgery, and late ruptures are cases that happen days<br />

to years after MVR. 1, 2 Because of the high propensity of<br />

the pseudoaneurysm to enlargement and rupture, immediate<br />

surgical management is inevitable. 3-5 We report a case<br />

with a late diagnosis of the LV pseudoaneurysm following<br />

previous MVR and coronary artery bypass graft surgery<br />

(CABG), which was diagnosed via two-dimensional and<br />

real-time three-dimensional transthoracic echocardiography<br />

(RT 3D TTE).<br />

Case Report<br />

A 74-year-old female with a history of coronary artery<br />

*<br />

Corresponding Author: Mohammad Sahebjam, Assistant Professor of Cardiology, Echocardiography Department, Tehran Heart Center, Jalal Al<br />

Ahmad and North Kargar Intersection, Tehran, Iran. 1411713138. Tel: +98 21 88029600. Fax: +98 21 88029731. E-mail: msahebjam@yahoo.com.<br />

188


Late Diagnosis of Large Left Ventricular Pseudoaneurysm after Mitral Valve ...<br />

disease was admitted to our hospital with a history of<br />

exertional dyspnea (function class II) during recent<br />

months. She had a past history of CABG and MVR surgery<br />

approximately 10 months prior to her admission.<br />

The postoperative echocardiography, performed one<br />

week after surgery, showed mild LV hypertrophy with an<br />

ejection fraction of 50%, a prosthetic MV with an acceptable<br />

gradient, and a normal right ventricle size with a good<br />

systolic function.<br />

On present admission, the patient’s pulse rate was 90 per<br />

minute and regular, and her blood pressure was 130/70 mm<br />

Hg. The patient had 1+ edema in her lower limbs. Auscultation<br />

revealed fine crackles in both lung bases and III/VI systolic<br />

murmur. An electrocardiogram (ECG) showed left bundle<br />

branch block and inverted T wave in V5-V6 leads.<br />

TTE showed a large echolucent space with a narrow neck<br />

(about 1.8 - 2 cm in diameter) on the basal posterolateral portion<br />

of the LV (near the prosthetic MV) and the diameter of the<br />

cavity was about 70 mm, suggestive of a large pseudoaneurysm<br />

in the basal posterolateral portion of the LV with a to-and-fro<br />

flow on color Doppler (Figures 1 and 2). RT 3D TTE (Vivid 7<br />

/ GE / RT 3D probe) was performed in order to better reveal<br />

the spatial relations between the pseudoaneurysm and the<br />

adjacent cardiovascular and thoracic structures and to better<br />

delineate the pseudoaneurysm borders before surgery. RT 3D<br />

TTE showed a large pseudoaneurysm (80 × 52 mm), parallel<br />

to the free wall of the LV (Figure 3). Its orifice was about 20<br />

mm and was very close to the MV annulus.<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

prosthetic MV was removed. In the posterolateral aspect of<br />

the LV just below the annulus of the MV, the large orifice<br />

of the pseudoaneurysm (about 20 mm in diameter) was<br />

found, which was closed with a pericardial patch (Figure<br />

4). After the closure of the defect, MVR with a bileaflet<br />

mechanical valve (St. Jude # 27) was performed and the<br />

patient was weaned from CPB without problems. Post-pump<br />

intraoperative transesophageal echocardiography confirmed<br />

the complete closure of the orifice of the pseudoaneurysm<br />

without a flow. The postoperative course was acceptable and<br />

the patient was discharged on 10 th postoperative day. TTE<br />

was performed before discharge and showed the prosthetic<br />

MV with an acceptable gradient, without paravalvular leak<br />

and a large echomixed cavity (7 × 3.8 cm) due to a previous<br />

pseudoaneurysm posterolateral to the LV, which was filled<br />

by a clot. At one year's follow-up, the patient was in good<br />

condition and was taking oral medications.<br />

Figure 2. Two-dimensional color Doppler echocardiography, showing<br />

a turbulent flow on the orifice of the pseudoaneurysm (arrow) and the<br />

pseudoaneurysm (asterisk)<br />

Figure 1. Two-dimensional echocardiography, showing the orifice of<br />

the pseudoaneurysm near the mitral valve sewing ring (arrow) and the<br />

pseudoaneurysm (asterisk)<br />

The patient underwent urgent surgery through median<br />

sternotomy using cardiopulmonary bypass. After mid<br />

sternotomy and moderate hypothermia (28 °C), the left<br />

atrium was opened. The prosthetic MV (St Jude # 27)<br />

remained intact, but the orifice of the LV pseudoaneurysm<br />

was unclear when visualized through the St Jude valve. The<br />

Figure 3. Three-dimensional echocardiography, showing the orifice of the<br />

pseudoaneurysm (arrow) and the pseudoaneurysm (asterisk)<br />

The Journal of Tehran University Heart Center189


The Journal of Tehran University Heart Center<br />

Mohammad Sahebjam et al.<br />

important for the operative approach plan.<br />

Conclusion<br />

The LV pseudoaneurysm is an important complication that<br />

may occur late after MVR and thus necessitates due heed on<br />

the part of cardiologists and cardiac surgeons.<br />

References<br />

Figure 4. The operation site. The black arrow points to the pseudoaneurysm<br />

orifice (PS) and the white arrow to the prosthetic mitral valve (MV)<br />

Discussion<br />

Rupture of the LV wall after MVR, albeit infrequent,<br />

is a fatal complication, especially in the delayed class. 4,<br />

6, 7<br />

The most frequent cause of the LV pseudoaneurysm is<br />

myocardial infarction secondary to atherosclerotic coronary<br />

artery disease, but many other causes may be responsible<br />

for the LV pseudoaneurysm such as trauma, infective<br />

endocarditis, inflammation, and cardiac surgery. 8 Congestive<br />

heart failure is the most common clinical presentation of the<br />

LV pseudoaneurysm but the rate of asymptomatic patients is<br />

more than 10%. 9 For the first time, Spellberg and O’Reilly<br />

reported post-MVR pseudoaneurysm, which was diagnosed<br />

via left ventriculography. 10 Two third of the LV ruptures are<br />

the early type, and the mortality rate of this category reaches<br />

near 50% despite early management. 10 Classification of<br />

the post-MVR pseudoaneurysm may be based on the time<br />

and location of the rupture. 1 Miller and co-workers also<br />

classified the LV pseudoaneurysm based on the location of<br />

the LV rupture and its relative location to the MV annulus. 10<br />

In patients with congestive heart failure symptoms, the<br />

mortality rate is high, particularly without surgery. One<br />

of the previous case series study reported 11 deaths in 35<br />

patients in non-operated cases with pseudoaneurysms. 9<br />

The initial technique for the diagnosis of postoperative<br />

complications is TTE. 1 Proper anatomic delineation is<br />

essential in order to plan appropriate therapy. Contrary<br />

to the post-myocardial infarction cases, these aneurysms<br />

following MVR tend to be sub-annular in location. 11<br />

Previously, pseudoaneurysm diagnosis was made by left<br />

ventriculography, while echocardiography and Cardiac<br />

Computed Tomography (CT) can be the other diagnostic<br />

approaches for these cases. 11<br />

The usage of RT 3D TTE in this case conferred better<br />

delineation of the pseudoaneurysm territory, which is<br />

1. Biyikoglu SF, Guray Y, Turkvatan A, Boyaci A, Katircioglu F.<br />

A serious complication late after mitral valve replacement: left<br />

ventricular rupture with pseudoaneurysm. J Am Soc Echocardiogr<br />

2008;21:1178.e1-3.<br />

2. Ono M, Wolf RK. Left ventricular pseudoaneurysm late after<br />

mitral valve replacement. Ann Thorac Surg 2002;73:1303-1305.<br />

3. Suda H, Ikeda K, Doi K, Shiraishi R, Furukawa K, Ito T. Successful<br />

repair of left ventricular pseudoaneurysm after mitral reoperation<br />

under hypothermic circulatory arrest. Jpn J Thorac Cardiovasc<br />

Surg 2003;51:18-20.<br />

4. Choi JB, Choi SH, Oh SK, Kim NH. Left ventricular<br />

pseudoaneurysm after coronary artery bypass and valve<br />

replacement for post-infarction mitral regurgitation. Tex Heart Inst<br />

J 2006;33:505-507.<br />

5. Nekkanti R, Nanda NC, Ansingkar KG, McGiffin DC.<br />

Transesophageal three-dimensional echocardiographic<br />

assessment of left ventricular pseudoaneurysm. Echocardiography<br />

2002;19:169-172.<br />

6. Kupari M, Verkkala K, Maamies T, Härtel G. Value of combined<br />

cross sectional and Doppler echocardiography in the detection of<br />

left ventricular pseudoaneurysm after mitral valve replacement. Br<br />

Heart J 1987;58:52-56.<br />

7. Honda K, Okamura Y, Nishiharu Y, Hayashi H. Patch repair<br />

of a giant left ventricular pseudoaneurysm after mitral valve<br />

replacement. Ann Thorac Surg 2011;91:1596-1597.<br />

8. Mahilmaran A, Nayar PG, Sheshadri M, Sudarsana G, Abraham<br />

KA. Left ventricular pseudoaneurysm caused by coronary spasm,<br />

myocardial infarction, and myocardial rupture. Tex Heart Inst J<br />

2002;29:122-125.<br />

9. Leitman M, Shmueli R, Stamler A, Krakover R, Hendler A,<br />

Vered Z. An unusual presentation of a large left ventricular<br />

pseudoaneurysm. Echocardiography 2006;23:403-404.<br />

10. Lanjewar C, Thakkar B, Kerkar P, Khandeparkar J. Submitral left<br />

ventricular Pseudoaneurysm after mitral valve replacement: early<br />

diagnosis and successful repair. Interact Cardiovasc Thorac Surg<br />

2007;6:505-507.<br />

11. Yeo TC, Malouf JF, Oh JK, Seward JB. Clinical profile and<br />

outcomein 52 patients with cardiac pseudoaneurysm. Ann Intern<br />

Med 1998;128:299-305.<br />

190


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Letter to the Editor<br />

A Memorandum of "World<br />

Heart Day 2012": Myocardial<br />

Infarction Mortality in Women<br />

in Birjand, 2008-2009<br />

September 29th, 2012 is entitled "World Heart Day" and<br />

the theme of the current year is "One World, One Heart, One<br />

Home, and Focus on Women and Children".<br />

Cardiovascular diseases are the most common causes of<br />

mortality in women and men. 1 Various studies have probed<br />

into the differences in the clinical symptoms, risk factors,<br />

and prognoses between male and female patients with acute<br />

myocardial infarction (AMI). 2<br />

One study on AMI patients in Birjand, Iran (2008-2009)<br />

reported the following results:<br />

Out of 471 patients, 136 (28.9%) cases were women<br />

and 335 (71.1%) were men. The mean age of the females<br />

and males was 64.6 ± 13.6 years and 61.4 ± 13.95 years,<br />

respectively; p value = 0.001.The most prevalent cardiac risk<br />

factors in the women and men were hypertension (54.4%<br />

in women, 32.2% in men; p value < 0.001), dyslipidemia<br />

(30.1% in women, 18.5% in men; p value = 0.006), diabetes<br />

mellitus (18.4% in women, 14.9% in men; p value = 0.35),<br />

and smoking (3.7% in women, 22.7% in men; p value =<br />

0.04).<br />

Table 1 shows that the consumption of Aspirin and beta<br />

blockers was lower in the women than in the men, but<br />

there was no other difference between the two genders with<br />

respect to other treatments. In-hospital mortality was 11.5%:<br />

8.4% in the men and 19.1% in the women (p value = 0.001).<br />

One-month mortality was 12.9%; it was significantly higher<br />

in the women than in the men (22.1% in women, 9.2% in<br />

men; p value ≤ 0.001). Finally, in-hospital mortality and<br />

one-month mortality after AMI were significantly higher in<br />

the women.<br />

Factors such as old age, higher prevalence of diabetes<br />

mellitus and hypertension, delayed referral of women to<br />

physicians, and difference in clinical symptoms can be play<br />

a decisive role in this regard. 3 Indeed, women do not tend to<br />

deem cardiovascular diseases as their most fatal disease and<br />

thus fail to take their risk factors into consideration; that is<br />

why cardiovascular diseases are diagnosed later in women<br />

and are more acute. It is, therefore, necessary that educational<br />

programs be devised to raise women's knowledge about<br />

these fatal diseases.<br />

Table 1. Comparison of drug therapy in patient with acute myocardial<br />

infarction (AMI) according to sex<br />

Drug<br />

Female<br />

n (%)<br />

Male<br />

n (%)<br />

P value<br />

Beta-blocker 77 (56.6) 222 (66.3) 0.04<br />

ASA 107 (78.7) 302 (90.1) 0.01<br />

Statins 74 (54.4) 213 (63.6) 0.06<br />

ACE-I 40 (29.4) 105 (31.3) 0.68<br />

Streptokinase 12 (8.8) 46 (13.7) 0.14<br />

Clopidogrel 27 (19.9) 90 (26.9) 0.11<br />

Nitrate 104 (76.5) 280 (83.6) 0.07<br />

ASA, Acetylsalicylic acid; ACE-I, Angiotensin-converting enzyme<br />

inhibitors<br />

References<br />

1. Kazemi T, Sharifzadeh GR, Zarban A, Fesharakinia A, Rezvani<br />

MR, Moezy SA. Risk factors for premature myocardial infarction:<br />

a matched case-control study. J Res Health Sci 201;11:77-82.<br />

2. Abbasi SH, Kassaian SE. Women and coronary artery disease. Part<br />

I: basic considerations. J Teh Univ Heart Ctr 2011;6:109-116.<br />

3. Hosseini SK, Soleimani A, Karimi AA, Sadeghian S, Darabian S,<br />

Abbasi SH, Ahmadi SH, Zoroufian A, Mahmoodian M, Abbasi<br />

A. Clinical features, management and in-hospital outcome of ST<br />

elevation myocardial infarction (STEMI) in young adults under 40<br />

years of age. Monaldi Arch Chest Dis 2009;72:71-76.<br />

Toba Kazemi, MD<br />

Associate Professor of Cardiology,<br />

Birjand Atherosclerosis and Coronary Artery Research Center,<br />

Department of Cardiology,<br />

Birjand University of Medical Sciences (BUMS),<br />

Valiassr Hospital, Ghafari Avenue,<br />

Birjand, Iran.<br />

9717964151.<br />

Tel: +98 56 14443001-9.<br />

Fax: +98 56 14433004.<br />

E-mail: drtooba.kazemi@gmail.com.<br />

Gholam Reza Sharifzadeh, MSc<br />

Epidemiologist,<br />

Birjand University of Medical Sciences (BUMS),<br />

Pasdaran Avenue,<br />

Birjand, Iran.<br />

9717964151.<br />

Tel: +98 56 14443001-9.<br />

Fax: +98 56 14433004.<br />

E-mail: rezamood@yahoo.com.<br />

The Journal of Tehran University Heart Center191


The Journal of Tehran University Heart Center<br />

Photo Clinic<br />

Ball in Chest<br />

Mohammad Bagher Rahim, MD, Mohammad Hossein Mandegar, MD,<br />

Farideh Roshanali, MD *<br />

Shariati General Hospital, Tehran University of Medical Sciences, Tehran, Iran.<br />

A 50-year-old male underwent a check-up for an insurance company. Chest X-ray revealed a large ball-like lesion in the<br />

posterior mediastinum. The finding was confirmed by computed tomography (CT) scan (Figure 1). The patient was referred<br />

to our center, where he had a successful mass removal surgery (Figure 2). Pathology identified the mass as neurinoma.<br />

A<br />

B<br />

Figure 1. Chest X-ray (A) and computed tomography (CT)<br />

scan (B), revealing a large ball-like lesion in the chest (arrows)<br />

Figure 2. The removed mass<br />

J Teh Univ Heart Ctr 2012;7(4):192<br />

This paper should be cited as: Rahim MB, Mandegar MH, Roshanali F. Ball in Chest. J Teh Univ Heart Ctr 2012;7(4):192.<br />

Keywords: Mass chest X-Ray • Thorax • Neoplasms<br />

*<br />

Corresponding Author: Farideh Roshanali, Shriati Hospital, North Kargar Street, Tehran, Iran. Tel: +98 912 3093151. Fax: +98 21 88797353.<br />

E-mail: farideh_roshanali@yahoo.com.<br />

192


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

INTERNATIONAL CARDIOVASCULAR SURGERY<br />

MEETINGS CALENDER (2012)<br />

Congress Time - Location Address<br />

Intermediate Skills in Cardiac Surgery<br />

2nd Leadership Course for Cardiovascular and<br />

Thoracic Surgeons<br />

31st Annual Winter Meeting of the Association of<br />

Physician Assistants in Cardiovascular Surgery<br />

(APACVS)<br />

STS/AATS Tech-Con 2012<br />

48th Annual Meeting of The Society of Thoracic<br />

Surgeons<br />

STS 48th Annual Meeting: Late-Breaking Clinical<br />

Trial Abstract Presentations<br />

ESAO Winter School 2012 "New Emerging<br />

Technology In Organ Failure" - Artificial Organs vs.<br />

Transplantation<br />

SCMR 15th Annual Scientific Sessions<br />

15th Comprehensive Review & Update of<br />

Perioperative Echo<br />

10-11 January 2012<br />

Birmingham<br />

United Kingdom<br />

16-20 January 2012<br />

Windsor<br />

United Kingdom<br />

26-29 January 2012<br />

Ft. Lauderdale, FL<br />

United States<br />

28-29 January 2012<br />

Fort Lauderdale, FL<br />

United States<br />

28 January-01 February 2012<br />

Fort Lauderdale, FL<br />

United States<br />

28 January-01 February 2012<br />

Fort Lauderdale, FL<br />

United States<br />

02-04 February 2012<br />

Catania, Sicily<br />

Italy<br />

02-05 February 2012<br />

Orlando, FL<br />

United States<br />

06-11 February 2012<br />

San Diego, CA<br />

United States<br />

http://ubis.rcsed.ac.uk/courses/<br />

coursedetails.aspxdiaryId=923<br />

http://www.eacts.org<br />

http://www.apacvs.org/<br />

http://www.sts.org/education-meetings/<br />

sts-annual-meeting/sts/aats-techcon-2011<br />

http://www.sts.org/education-meetings/stsannual-meeting<br />

http://www.sts.org/education-meetings/<br />

sts-annual-meeting/abstract-submission<br />

http://www.esaowinterschool2012.it<br />

http://www.scmr.org/meetings.html<br />

http://www.scahq.org/sca3/events/2012/<br />

echo/<br />

The Journal of Tehran University Heart Center


The Journal of Tehran University Heart Center<br />

Congress Time - Location Address<br />

1st IACTS-EACTS Joint Worksop<br />

The 22nd Annual Scientific Meeting SSRCTS<br />

58th Annual Meeting of Indian Association of<br />

Cardiovascular & Thoracic Surgeons (IACTS)<br />

41th Annual Meeting of the German Society for<br />

Thoracic - Cardiovascular Surgery GSTCVS<br />

ESTS School of Thoracic Surgery (Practical Course<br />

in the Laboratory)<br />

Course of Minimal Invasive Techniques in Adult<br />

Cardiac Surgery<br />

12th Annual International Symposium on Congenital<br />

Heart Disease<br />

The 32nd Annual Cardiothoracic Surgery<br />

Symposium (CREF 2012)<br />

8th International Congress of Update In Cardiology<br />

And Cardiovascular Surgery<br />

30th Cardiovascular Surgical Symposium<br />

Re-Evolution Summit III: Minimally Invasive<br />

Cardiac Surgery (MICS) and Hybrid Strategies<br />

Mitral Conclave Course: AATS Postgraduate Course<br />

17th Annual Update on Cardiopulmonary Bypass<br />

ESTS School of Thoracic Surgery (Theoretical<br />

Course)<br />

Florida Valve 2012 "Cardiovascular Practice<br />

Transformation"<br />

Houston Aortic Symposium: Frontiers in<br />

Cardiovascular Diseases, The Fifth in the Series<br />

08-09 February 2012<br />

Kolkata, West Bengal<br />

India<br />

09-11 February 2012<br />

Geilo<br />

Norway<br />

10-12 February 2012<br />

Kolkata, West Bengal<br />

India<br />

12-15 February 2012<br />

Freiburg<br />

Germany<br />

13-14 February 2012<br />

Paris<br />

France<br />

13-15 February 2012<br />

Nieuwegein<br />

Netherlands<br />

18-21 February 2012<br />

St. Petersburg, FL<br />

United States<br />

29 February-04 March 2012<br />

San Diego, CA<br />

United States<br />

01-04 March 2012<br />

Antalya<br />

Turkey<br />

03-10 March 2012<br />

Zuers/Arlberg<br />

Austria<br />

08-10 March 2012<br />

Houston, TX<br />

United States<br />

09-11 March 2012<br />

Bali<br />

Indonesia<br />

11-16 March 2012<br />

Snowmass Village, CO<br />

United States<br />

14-18 March 2012<br />

Antalya<br />

Turkey<br />

21-23 March 2012<br />

Orlando, FL<br />

United States<br />

22-24 March 2012<br />

Houston, TX<br />

United States<br />

http://www.iacts2012.com<br />

http://www.ssrcts.org/<br />

http://www.iacts2012.com<br />

http://www.dgthg-jahrestagung.de/en<br />

http://www.estsschool.org<br />

http://www.eacts.org<br />

http://www.allkids.org/conferences<br />

http://www.amainc.com/cref_<br />

cardiothoracic.html<br />

http://www.uccs2012.org<br />

http://www.surgery-zurs.at<br />

http://www.methodisthealth.com/basic.<br />

cfmid=37173<br />

http://www.ascvtsbali2012.org/program.<br />

html<br />

http://www.scahq.org/sca3/events/2012/<br />

cpb/<br />

http://www.estsschool.org<br />

http://www.floridavalvesymposium.com<br />

http://www.promedicacme.com/meeting/<br />

Houston-Aortic-Symposium-Frontiersin-73.html


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Congress Time - Location Address<br />

AmSECT's 50th International Conference Meeting<br />

The 10th Annual International Nuss Pectus<br />

Excavatum and Carinatum Lecture Series<br />

Minimally Invasive Robotic Association (MIRA):<br />

7th International Congress<br />

Valves in the Heart of the Big Apple VII:<br />

Evaluation & Management of Valvular Heart<br />

Disease 2012<br />

34th International Symposium CX Charing Cross<br />

Controversies Challenge Consensus - Vascular &<br />

Endovascular Controviersies Update<br />

Association of Cardiothoracic Anesthetists & Society for<br />

Cardiothoracic Surgery in Great Britain and Ireland -<br />

2012 Annual Meeting and Cardiothoracic Forum<br />

Arteriosclerosis, Thrombosis and Vascular Biology<br />

(ATVB) 2012 Scientific Sessions<br />

32nd Annual Meeting & Scientific Sessions of<br />

The International Society of Heart and Lung<br />

Transplantation (ISHLT)<br />

SNM's 3rd Multimodality Cardiovascular Molecular<br />

Imaging Symposium<br />

61st Annual Meeting of the European Society for<br />

Cardiovascular Surgery (ESCVS)<br />

Aortic Symposium 2012<br />

Annual Thoracic Anesthesia Symposium<br />

34th Annual Meeting of the Society of Cardiovascular<br />

Anesthesiologists & Workshops<br />

92nd Annual Meeting - American Association for<br />

Thoracic Surgery<br />

5th Biennial Meeting on Heart Valve Biology and<br />

Tissue Engineering<br />

International Society for Minimally Invasive Cardiac<br />

Surgery 2012 ISMICS Annual Meeting<br />

28-31 March 2012<br />

Orlando, Florida<br />

United States<br />

29-30 March 2012<br />

Norfolk, VA<br />

United States<br />

11-13 April 2012<br />

Baltimore, MD<br />

United States<br />

12-14 April 2012<br />

New York City, NY<br />

United States<br />

14-17 April 2012<br />

London<br />

United Kingdom<br />

18-20 April 2012<br />

Manchester<br />

United Kingdom<br />

18-20 April 2012<br />

Chicago, IL<br />

United States<br />

18-21April 2012<br />

Prague<br />

Czech Republic<br />

19-21 April 2012,<br />

Bethesda, Maryland<br />

United States<br />

25-28 April 2012<br />

Dubrovnik<br />

Croatia<br />

26-27 April 2012<br />

New York, NY<br />

United States<br />

27-28 April 2012<br />

Boston, MA<br />

United States<br />

28 April-02 May 2012<br />

Boston, MA<br />

United States<br />

28 April-02 May 2012<br />

San Francisco, CA<br />

United States<br />

18-20 May 2012<br />

Mykonos<br />

Greece<br />

30 May-02 June 2012<br />

Los Angeles, CA<br />

United States<br />

http://amsect.societyhq.com/meetings/<br />

international/2012/abstracts/<br />

http://www.chkd.org/pectus<br />

http://www.mirasurgery.org/events.php<br />

http://www.heartvalveconference.com/<br />

http://www.cxvascular.com/<br />

cxsymposium<br />

http://www.scts.org/<br />

http://my.americanheart.org/professional/<br />

Sessions/ATVB/ATVB_UCM_316902_<br />

SubHomePage.jsp<br />

http://www.ishlt.org/meetings/<br />

annualMeeting.asp<br />

www.snm.org/cvmi2012<br />

http://www.escvs.com<br />

http://www.aats.org/aortic/<br />

http://www.scahq.org/sca3/events/2012/<br />

thoracic/<br />

http://www.scahq.org/sca3/events/2012/<br />

annual/<br />

http://www.aats.org/annualmeeting<br />

http://greeceheartvalve.com/home.asp<br />

http://www.ismics.org/<br />

The Journal of Tehran University Heart Center


The Journal of Tehran University Heart Center<br />

Congress Time - Location Address<br />

47th Congress of the European Society for Surgical<br />

Research ESSR 2012<br />

6th Congress of the Asian Society of Cardiovascular<br />

Imaging (ASCI)<br />

22nd congress of the world society of cardiothoracic<br />

surgeons<br />

20th European Conference on General Thoracic<br />

Surgery<br />

8th International Conference Pediatric Mechanical<br />

Circulatory Support Systems and Pediatric<br />

Cardiopulmonary Perfusion<br />

American Society of Arificial Internal Organs -<br />

ASAIO 58th Annual Conference<br />

6th Annual The New Orleans Conference: Practices<br />

in Cardiac Surgery and Extracorporeal Technologies<br />

The 3rd Beijing International Symposium and<br />

Workshop on Robotic Cardiac Surgery (BISROCS<br />

2012)<br />

4th Joint Conference of Scandinavian Association<br />

for Thoracic Surgery<br />

Georgia Health Sciences University Cardiac<br />

Conference<br />

ESTS School of Thoracic Surgery (Practical Course<br />

in the Laboratory)<br />

26th EACTS Annual Meeting<br />

National Meeting of Turkish Society of<br />

Cardiovascular Surgery<br />

Second International Joint Meeting on Thoracic<br />

Surgery, Barcelona 2012<br />

06-09 June 2012<br />

Lille<br />

France<br />

07-09 June 2012<br />

Bangkok<br />

Thailand<br />

09-13 June 2012<br />

Vancouver<br />

Canada<br />

10-13 June 2012<br />

Essen<br />

Germany<br />

13-16 June 2012<br />

Istanbul<br />

Turkey<br />

14-16 June 2012<br />

San Francisco, CA<br />

United States<br />

27-30 June 2012<br />

New Orleans, LA<br />

United States<br />

14-16 July 2012<br />

Beijing<br />

China<br />

16-18 August 2012<br />

Vilnius<br />

Lithuania<br />

21-23 September 2012<br />

Braselton, GA<br />

United States<br />

24-25 September 2012<br />

Paris<br />

France<br />

27-31 October 2012<br />

Barcelona<br />

Spain<br />

8-11 November 2012<br />

Antalya<br />

Turkey<br />

14-16 November 2012<br />

Barcelona<br />

Spain<br />

http://www.essr2012.com/<br />

http://www.asci2012.org<br />

www.wscts.org<br />

www.ests.org<br />

http://pennstatehershey.org/web/pedscpb/<br />

home<br />

http://www.asaio.com<br />

http://www.TheNewOrleansConference.<br />

com<br />

http://www.roboticsurg301.net<br />

http://www.scandinavian-ats.org/<br />

http://www.<br />

georgiahealthcardiacconference.com/<br />

http://www.estsschool.org<br />

http://www.eacts.org<br />

http://www.tkdcd.org<br />

http://thoracicsurgery2012.org/


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

INTERNATIONAL CARDIOVASCULAR MEETING<br />

AND CONGRESSES CALENDER (2012)<br />

Title Location Start Date End Date<br />

3rd Asia PCR/ SingLive 2012<br />

Singapore<br />

Republic of Singapore<br />

12 January 2012<br />

14 January 2012<br />

ISET 2012: International Symposium on Endovascular<br />

Therapy<br />

Miami Beach, Florida<br />

United States<br />

15 January 2012<br />

19 January 2012<br />

44th Ten-Day International Teaching Seminar on<br />

Cardiovascular Disease Epidemiology and Prevention<br />

Cape Town<br />

South Africa<br />

15 January 2012<br />

27 January 2012<br />

Ninth Gulf Heart Association Conference<br />

Muscat<br />

Oman<br />

18 January 2012<br />

21 January 2012<br />

22nd Annual Echo Hawaii 2012<br />

Big Island, HI<br />

United States<br />

23 January 2012<br />

27 January 2012<br />

6th Middle East Cardiovascular Congress & IVth<br />

International Congress of Cardiovascular Imaging<br />

Dubai<br />

UAE<br />

24 January 2012<br />

27 January 2012<br />

33rd Annual Seminar of The American Academy of<br />

Cardiovascular Perfusion<br />

New Orleans, Louisiana<br />

United States<br />

26 January 2012<br />

29 January 2012<br />

19th Annual Arrhythmias & the Heart: A Cardiovascular<br />

Update<br />

Kauai<br />

Hawaii<br />

30 January 2012<br />

03 February 2012<br />

31st Annual Scientific Meeting of the Belgian Society of<br />

Cardiology<br />

Brussels<br />

Belgium<br />

02 February 2012<br />

03 February 2012<br />

International Congress on Personalized Medicine: Up<br />

Close and Personalized (UPCP 2012)<br />

Florence<br />

Italy<br />

02 February 2012<br />

05 February 2012<br />

APCHF 2012<br />

6th Asian Pacific Congress of Heart Failure<br />

Chiang Mai<br />

Thailand<br />

03 February 2012<br />

05 February 2012<br />

Cardiovascular Research Technologies (CRT) 2012<br />

Washington DC<br />

United States<br />

04 February 2012<br />

07 February 2012<br />

The Journal of Tehran University Heart Center


The Journal of Tehran University Heart Center<br />

Title Location Start Date End Date<br />

Vascular Care 2012: New Technologies and Emerging<br />

Therapies<br />

Truckee, CA<br />

United States<br />

05 February 2012<br />

08 February 2012<br />

37th Annual Cardiovascular Conference at Snowbird<br />

Snowbird, UT<br />

United States<br />

05 February 2012<br />

08 February 2012<br />

Gulf Heart Association & Saudi Heart Association Annual<br />

Conference<br />

Riyadh<br />

Saudi Arabia<br />

06 February 2012<br />

09 February 2012<br />

10th International Kawasaki Disease Symposium<br />

Kyoto<br />

Japan<br />

07 February 2012<br />

10 February 2012<br />

13th International Forum for the Evaluation of<br />

Cardiovascular Care<br />

Nice<br />

France<br />

09 February 2012<br />

11 February 2012<br />

Icon 2012- International Congress for Endovascular<br />

Specialists<br />

Scottsdale, Arizona<br />

United States<br />

12 February 2012<br />

16 February 2012<br />

12th Annual International Symposium on Congenital Heart<br />

Disease (CHD)<br />

Florida<br />

United States<br />

18 February 2012<br />

18 February 2012<br />

11th Genoa Meeting on Hypertension, Diabetes and Renal<br />

Disease<br />

Genoa<br />

Italy<br />

23 February 2012<br />

25 February 2012<br />

2nd Annual Case Based Approach to Controversies in<br />

Cardiovascular Disease<br />

Dubai<br />

UAE<br />

23 February 2012<br />

25 February 2012<br />

Cardiovascular Disease Prevention 2012: Tenth Annual<br />

Comprehensive Symposium<br />

Miami Beach, Florida<br />

United States<br />

23 February 2012<br />

26 February 2012<br />

17th Annual Cardiology at Cancun: Topics in Clinical<br />

Cardiology: Focus on Heart Failure<br />

Cancun<br />

Mexico<br />

27 February 2012<br />

02 March 2012<br />

NCVH Latin America<br />

An International Approach to Lower Limb Interventions<br />

San Jose<br />

Costa Rica<br />

28 February 2012<br />

01 March 2012<br />

The Future of Genomic Medicine V<br />

La Jolla, California<br />

United States<br />

01 March 2012<br />

02 March 2012<br />

International Conference on Heart & Brain<br />

Paris<br />

France<br />

01 March 2012<br />

03 March 2012<br />

8th International Congress of Update in Cardiology and<br />

Cardiovascular Surgery<br />

Antalya<br />

Turkey<br />

01 March 2012<br />

04 March 2012<br />

2012 Hypertension Congress<br />

Cape Town<br />

South Africa<br />

03 March 2012<br />

05 March 2012<br />

Interventional Cardiology 2012: 27th Annual International<br />

Symposium<br />

Snowmass Village, CO<br />

United States<br />

04 March 2012<br />

09 March 2012<br />

10th National Congress of the Italian Society for<br />

Cardiovascular Prevention SIPREC<br />

Napoli<br />

Italy<br />

08 March 2012<br />

10 March 2012<br />

9th Mediterranean Meeting on Hypertension and<br />

Atherosclerosis<br />

Antalya<br />

Turkey<br />

14 March 2012<br />

18 March 2012<br />

12th Annual Spring Meeting on Cardiovascular Nursing<br />

Copenhagen<br />

Denmark<br />

16 March 2012<br />

17 March 2012<br />

76th Annual Scientific Meeting of the Japanese Circulation<br />

Society<br />

Fukuoka<br />

Japan<br />

16 March 2012<br />

18 March 2012<br />

The Houston Aortic Symposium - Frontiers in Cardiovascular<br />

Diseases: The Fifth in the Series<br />

Houston, TX<br />

United States<br />

22 March 2012<br />

24 March 2012


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Title Location Start Date End Date<br />

61st Annual American College of Cardiology Scientific<br />

Sessions<br />

Chicago<br />

United States<br />

24 March 2012<br />

27 March 2012<br />

Frontiers in CardioVascular Biology 2012<br />

London<br />

United Kingdom<br />

30 March 2012<br />

01 April 2012<br />

Cardiology in the Young 2012<br />

London<br />

United Kingdom<br />

02 April 2012<br />

04 April 2012<br />

German Society of Cardiology 78th Annual Meeting 2012<br />

(DGK 2012)<br />

Manheim<br />

Germany<br />

11 April 2012<br />

14 April 2012<br />

34th Charing Cross International Symposium<br />

London<br />

United Kingdom<br />

14 April 2012<br />

17 April 2012<br />

Arteriosclerosis, Thrombosis and Vascular Biology 2012<br />

Scientific Sessions<br />

Chicago, IL<br />

United States<br />

18 April 2012<br />

20 April 2012<br />

World Congress of Cardiology Scientific Sessions<br />

Dubai<br />

UAE<br />

18 April 2012<br />

21 April 2012<br />

18th Annual Interventional Cardiology Fellows Course<br />

Miami, FL<br />

United States<br />

19 April 2012<br />

22 April 2012<br />

7th Clinical Update on Cardiac MRI & CT<br />

Cannes<br />

France<br />

20 April 2012<br />

22 April 2012<br />

Portuguese Society of Cardiology 33rd Congress 2012 (SPC<br />

2012)<br />

Vilamoura<br />

Portugal<br />

22 April 2012<br />

24 April 2012<br />

Angioplasty Summit Transcatheter Cardiovascular<br />

Therapeutics Asia Pacific 2012<br />

Seoul<br />

Korea<br />

25 April 2012<br />

27 April 2012<br />

22nd European Meeting On Hypertension and<br />

Cardiovascular Protection<br />

London<br />

United Kingdom<br />

26 April 2012<br />

29 April 2012<br />

Concepts in Contemporary Cardiovascular Medicine 2012<br />

Houston, TX<br />

United States<br />

01 May 2012<br />

04 May 2012<br />

EuroPRevent 2012<br />

Dublin<br />

Ireland<br />

03 May 2012<br />

05 May 2012<br />

APSC Intervention and Imaging 2012 Subspecialty Congress<br />

Taipei<br />

Taiwan<br />

04 May 2012<br />

06 May 2012<br />

Taiwan Society of Cardiology<br />

Annual Convention & Scientific Sessions<br />

Taipei<br />

Taiwan<br />

05 May 2012<br />

06 May 2012<br />

Quality of Care and Outcomes Research in Cardiovascular<br />

Disease and Stroke 2012 Scientific Sessions<br />

Atlanta, GA<br />

United States<br />

9 May 2012<br />

9 May 2012<br />

SCAI 2012 Scientific Sessions<br />

Las Vegas<br />

United States<br />

09 May 2012<br />

12 May 2012<br />

Heart Rythm 2012<br />

Boston<br />

United States<br />

09 May 2012<br />

12 May 2012<br />

EuroPCR 2012<br />

Paris<br />

France<br />

15 May 2012<br />

18 May 2012<br />

Metabolic Syndrome, Type II Diabetes and Atherosclerosis<br />

Marrakesh<br />

Morocco<br />

16 May 2012<br />

20 May 2012<br />

2nd International Congress on Cardiac Problems in<br />

Pregnancy (CPP 2012)<br />

Berlin<br />

Germany<br />

17 May 2012<br />

20 May 2012<br />

The Journal of Tehran University Heart Center


The Journal of Tehran University Heart Center<br />

Title Location Start Date End Date<br />

Heart Failure Congress 2012<br />

Belgrade<br />

Serbia<br />

19 May 2012<br />

22 May 2012<br />

EAS 2012, the 80th European Atherosclerosis Society<br />

Congress<br />

Milan<br />

Italy<br />

25 May 2012<br />

28 May 2012<br />

New Cardiovascular Horizons 13th Annual Conference<br />

New Orleans<br />

United States<br />

06 June 2012<br />

09 June 2012<br />

6th Congress of the Asian Society of Cardiovascular<br />

Imaging<br />

Bangkok<br />

Thailand<br />

07 June 2012<br />

09 June 2012<br />

15th Annual 2011 Live Symposium of Complex Coronary<br />

and Vascular Cases<br />

C3 Complex Cardiovascular Catheter Therapeutics-<br />

Advanced Endovascular and Coronary Intervention<br />

Global Summit 2012<br />

New York, NY<br />

United States<br />

Orlando, FL<br />

United States<br />

13 June 2012<br />

19 June 2012<br />

16 June 2012<br />

23 June 2012<br />

The iCi 2012 - imaging in cardiovascular interventions<br />

Frankfurt<br />

Germany<br />

27 June 2012<br />

27 June 2012<br />

CSI 2012- European congress on congenital and structural<br />

interventions<br />

Frankfurt<br />

Germany<br />

28 June 2012<br />

30 June 2012<br />

The 9th Tunisian European Days of cardiology practice<br />

Hammam Sousse<br />

Tunisia<br />

28 June 2012<br />

30 June 2012<br />

25th World Congress of the International Union of<br />

Angiology<br />

Prague<br />

Czech Republic<br />

01 July 2012<br />

05 July 2012<br />

15th Annual Cardiothoracic Update and TEE Board<br />

Review<br />

South Carolina<br />

United States<br />

05 July 2012<br />

08 July 2012<br />

International Society for the Study of Hypertension in<br />

Pregnancy 18th World Congress 2012 (ISSHP 2012)<br />

Geneva<br />

Switzerland<br />

09 July 2012<br />

12 July 2012<br />

19th ASEAN Federation of Cardiology Congress 2012<br />

Singapore<br />

Republic of Singapore<br />

13 July 2012<br />

15 July 2012<br />

Basic Cardiovascular Sciences 2012 Scientific Sessions<br />

New Orleans, LA<br />

United States<br />

23 July 2012<br />

26 July 2012<br />

Ten-Day Seminar on the Epidemiology and Prevention<br />

of Cardiovascular Disease<br />

Tahoe City, CA<br />

United States<br />

22 July 2012<br />

03 August 2012<br />

6th Annual Australian and New Zealand Endovascular<br />

Therapies Meeting 2012 (Anzet 2012)<br />

Brisbane<br />

Australia<br />

15 August 2012<br />

16 August 2012<br />

60th Annual Scientific Meeting of the Cardiac Society of<br />

Australia and New Zealand (CSANZ 2012)<br />

Brisbane<br />

Australia<br />

16 August 2012<br />

19 August 2012<br />

European Society of Cardiology Congress 2012<br />

Munich<br />

Germany<br />

25 August 2012<br />

29 August 2012<br />

CIRSE 2012 /Cardiovascular and Interventional<br />

Radiological Society of Europe<br />

Lisbon<br />

Portugal<br />

15 September 2012<br />

19 September 2012<br />

High Blood Pressure Research 2012 Scientific Sessions<br />

Washington DC<br />

United States<br />

19 September 2012<br />

22 September 2012<br />

Hypertension Sydney 2012<br />

Sydney<br />

Australia<br />

29 September 2012<br />

04 October 2012<br />

28th Annual Echocardiography in Pediatric and Adult<br />

Congenital Heart Disease<br />

Rochester, MN<br />

United States<br />

07 October 2012<br />

10 October 2012


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

Information for Authors<br />

The first three consecutive issues of "The Journal of Tehran University Heart Center" were published under the title of<br />

"The Journal of Tehran Heart Center" with ISSN: 1735-5370. From the fourth issue onward, however, the journal has been<br />

entitled ‘’The Journal of Tehran University Heart Center" with ISSN:1735-8620.<br />

Scope of the journal<br />

"The Journal of Tehran University Heart Center" aims to publish the highest quality material, both clinical and scientific,<br />

on all aspects of Cardiovascular Medicine. It includes articles related to research findings, technical evaluations, and reviews.<br />

In addition, it provides a forum for the exchange of information on all aspects of Cardiovascular Medicine, including<br />

educational issues. "The journal of Tehran University Heart Center" is an international, English language, peer reviewed<br />

journal concerned with Cardiovascular Medicine. It is an official journal of the Cardiovascular Research Center of the Tehran<br />

University of Medical Sciences (in collaboration with the Iranian Society of Cardiac Surgeons) and is published quarterly.<br />

Papers submitted to this journal which do not adhere to the Instructions for Authors will be returned for appropriate revision<br />

to be in line with the Instructions for Authors. They may then be resubmitted. Submission of an article implies that the work<br />

described has not been published previously (except in the form of an abstract or as part of a published lecture or academic<br />

thesis), that it is not under consideration for publication elsewhere, that its publication is approved by all Authors and tacitly<br />

or explicitly by the responsible authorities where the work was carried out, and that, if accepted, it will not be published<br />

elsewhere in the same form, in English or in any other language, without the written consent of the publisher.<br />

Article Categories<br />

The Journal of Tehran University Heart Center” accepts the following categories of articles:”<br />

Guest Editorial<br />

Original Article<br />

Clinical and pre-clinical papers based on either normal subjects or patients and the result of cardiovascular pre-clinical<br />

research will be Considered for publication provided they have an obvious clinical relevance.<br />

Brief communication<br />

Case report<br />

Review Article<br />

"The Journal of Tehran University Heart Center" publishes a limited number of scholarly, comprehensive reviews whose<br />

aims are to summarize and critically evaluate research in the field addressed and identify future implications. Reviews should<br />

not exceed 5000 words.<br />

Letter to editor<br />

Letters to the editor must not exceed 500 words and should focus on a specific article published in "The Journal of<br />

Tehran University Heart Center" within the preceding 12 weeks. No original data may be included. Authors will receive prepublication<br />

proofs, and the authors of the article cited invited to reply.<br />

Submission of manuscripts<br />

Four double spaced copies on 8 1/2 × 11 in. paper should be sent to:<br />

Dr. A. Karimi,<br />

Editor in Chief,<br />

"The Journal of Tehran University Heart Center",<br />

Tehran Heart Center,<br />

North Kargar Street,<br />

The Journal of Tehran University Heart Center


The Journal of Tehran University Heart Center<br />

Tehran, Iran.<br />

1411713138.<br />

Photocopies or good reproductions of illustrations are acceptable only on the spare copies. Included also should be a set<br />

of the electronic files of the manuscript on floppy - disk or CD-ROM. For preparation of electronic files, see the instructions<br />

herein below.<br />

Also, manuscripts can be submitted electronically via the journal’s website: http://jthc.tums.ac.ir. On-line submission<br />

allows the manuscript to be handled in electronic forms throughout the review process.<br />

Review of manuscripts<br />

All manuscripts correctly submitted to will first be reviewed by the Editors. Some manuscripts will be returned to authors<br />

at this stage if the paper is deemed inappropriate for publication in “The Journal of Tehran University Heart Center”, if the<br />

paper does not meet submission requirements, or if the paper is not deemed to have a sufficiently high priority. All papers<br />

considered suitable by the Editors to progress further in the review process will undergo appropriate peer review and all<br />

papers provisionally accepted for publication will undergo a detailed statistical review.<br />

Preparation of manuscripts<br />

All submitted manuscripts must not exceed 5000 words, including References, Figure Legends and Tables. The number of<br />

Tables, Figures and References should be appropriate to the manuscript content and should not be excessive. Authors should<br />

comply with the manuscript formatting and the ethical conventions of the “Uniform Requirements for Manuscripts Submitted<br />

to Biomedical Journals” issued by the International Committee of Medical Journal Editors ( http://www.icmje.org ).<br />

Style and spelling<br />

Authors whose first language is not English are requested to have their manuscripts checked carefully before submission.<br />

This will help expedite the review process and avoid confusion. Abbreviations of standard SI units of measurement only<br />

should be used.<br />

Declaration of Helsinki<br />

The Authors should state that their study complies with the Declaration of Helsinki that the locally appointed ethics<br />

committee has approved the research protocol and that informed consent has been obtained from the subjects (or their<br />

guardians).<br />

Clinical trials<br />

Authors should comply with the clinical trial registration statement from the ICMJE. More information can be found at<br />

www.icmje.org. Clinical trial reports should also comply with the Consolidated Standards of Reporting Trials (CONSORT)<br />

and include a flow diagram presenting the enrollment, intervention allocation, follow-up, and data analysis with number of<br />

subjects for each (www.consort-statement.org). Please also refer specifically to the CONSORT Checklist of items to include<br />

when reporting a randomized clinical trial.<br />

Section of the manuscripts<br />

Original articles should be divided into the following sections: (1) Title page, (2) Abstract and Keywords, (3) Introduction,<br />

(4) Methods, (5) Results, (6) Discussion, (7) Conclusion, (8) Acknowledgements, (9) References, (10) Figure legends,<br />

(11) Tables, (12) Figures.<br />

General format<br />

Prepare your manuscript text using a word processing package. Submissions of text in the form of PDF files are not


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

permitted. Manuscripts should be double-spaced, including text, tables, legends and references. Number each page. Please<br />

avoid footnotes; use instead, and as sparingly as possible, parenthesis within brackets. Enter text in the style and order of<br />

the Journal. Type references in the correct order and style of the journal. Type unjustified, without hyphenation, except for<br />

compound words. Type headings in the style of the journal. Use the TAB key once for paragraph indents. Where possible use<br />

Times New Roman for the text font and Symbol for the Greek and special characters. Use the word processing formatting<br />

features to indicate Bold, Italic, Greek, Maths, Superscript and subscript characters. Clearly identify unusual symbols and<br />

Greek letters. Differentiate between the letter o and zero, and the letters I and i and the number 1. Mark the approximate<br />

position of each figure and table. Check the final copy of your paper carefully, as any spelling mistakes and errors may be<br />

translated into the typeset version.<br />

Title page<br />

The title page should include the following: (1) the title, (2) the name (s) of authors and their highest degree ( no more<br />

than 12 authors are acceptable), (3) the institution (s) where work was performed, (4) institution, and location of all authors,<br />

(5) the address, telephone number, fax number and e-mail address of the corresponding author.<br />

Abstract<br />

All abstracts may not contain more than 250 words and should also be submitted as a separate file. The abstract should be<br />

formatted with the following heading: (1) Background, (2) Methods, (3) Results, (4) Conclusion.<br />

A maximum of six Keywords may be submitted.<br />

Figures<br />

The review process will not begin until all figures are received. Figures should be limited to the number necessary for<br />

clarity and must not duplicate data given in tables or in the text. They must be suitable for high quality reproduction and<br />

should be submitted in the desired final printed size so that reduction can be avoided. Figures should be no larger than 125<br />

(height)×180 (width) mm (5×7 inches) and should be submitted in a separate file from that of the manuscript.<br />

Electronic submission of figures<br />

Figures should be saved in TIFF format at a resolution of at least 300 pixels per inch at the final printed size for colour<br />

figures and photographs, and 1200 pixels per inch for black and white line drawings. Although some other formats can be<br />

translated into TIFF format by the publisher, the conversion may alter the tones, resolution and contrast of the image. Digital<br />

colour art should be submitted in CMYK rather than RGB format, as the printing process requires colours to be separated<br />

into CMYK and this conversion can alter the intensity and brightness of colours. Therefore authors should be satisfied with<br />

the colours in CMYK (both on screen and when printed) before submission. Please also keep in mind that colours can appear<br />

differently on different screens and printers. Failure to follow these guides could result in complications and delays.<br />

Photographs: Photographs should be of sufficiently high quality with respect to detail, contrast and fineness of grain to<br />

withstand the inevitable loss of contrast and detail inherent in the printing process. Please indicate the magnification by a rule<br />

on the photograph. Colour figures: There is a special charge for the inclusion of colour figures. Figure legends: These should<br />

be on a separate, numbered manuscript sheet grouped under the heading “Legends” on a separate sheet of the manuscript<br />

after the References. Define all symbols and abbreviations used in the figure. All abbreviations and should be redefined in<br />

the legend.<br />

Tables<br />

Tables should be typed with double spacing, but minimizing redundant space and each should be placed on a separate<br />

sheet. Tables should be submitted, wherever possible, in portraits, as opposed to landscape, layout. Each Table should be<br />

numbered in sequence using Arabic numerals. Tables should also have a title above and an explanatory footnote below. All<br />

abbreviations and should be redefined in the Footnote.<br />

The Journal of Tehran University Heart Center


The Journal of Tehran University Heart Center<br />

Acknowledgements<br />

All sources of funding and support, and substantive contributions of individuals, should be noted in the Acknowledgements,<br />

positioned before the list of references.<br />

Reference format<br />

Number references sequentially and use Arabic number in superscript to cite the reference in the text. All references<br />

should be compiled at the end of the article in the Vancouver style. Complete information should be given for each reference<br />

including the title of the article, abbreviated journal title and page numbers. All authors should be listed. Personal communications;<br />

manuscripts in preparation and other unpublished data should not be cited in the reference list but may be mentioned<br />

in parentheses in the text. Authors should get permission from the source to cite unpublished data.<br />

Titles of journals should be abbreviated in accordance with Index Medicus (see list printed annually in the January issue<br />

of Index Medicus). If a journal is not listed in Index Medicus then its name should be written out in full.<br />

Article citation example:<br />

Journal citation example: 1. Schroeder S, Baumbach A, Mahrholdt H. The impact of untreated coronary dissections on the<br />

acute and long-term outcome after intravascular ultrasound guided PTCA. Eur Heart J 2000;21:137-145.<br />

Chapter citation example: 2. Nichols WW, O’Rourke MF. Aging, high blood pressure and disease in humans. In: Arnold<br />

E, ed. McDonald’s Blood Flow in Arteries: Theoretical, Experimental and Clinical Principles. 3rd ed. London/Melbourne/<br />

Auckland: Lea and Febiger; 1990. p. 398-420.<br />

Webpage citation example: 3. Panteghini M. Recommendations on use of biochemical markers in acute coronary syndrome:<br />

IFCC proposals. eJIFCC 14. http://www.ifcc.org/ejifcc/vol14no2/1402062003014n.htm (28 May 2004). Where the<br />

date in parenthesis refers to the access date.<br />

Statistics<br />

All manuscripts selected for publication will be reviewed for the appropriateness and accuracy of the statistical methods<br />

used and the interpretation of statistical results. All papers submitted should provide in their Methods section a subsection<br />

detailing the statistical methods, including the specific method used to summarize the data, the methods used to test their<br />

hypothesis testing and (if any) the level of significance used for hypothesis testing.<br />

Conflict of interest<br />

At submission, the editors require authors to disclose any financial association that might pose a conflict of interest in<br />

connection with the submitted article. All sources of funding for the work should be acknowledged in a footnote on the title<br />

page and in the Acknowledgements within the manuscript, as should all the institutional affiliations of the authors (including<br />

corporate appointments). Other kinds of associations, such as consultancies, stock ownership or other equity interest<br />

or patent-licensing arrangements should be disclosed to the editors in the cover letter at the time of the of submission. If no<br />

conflict of interest exists, please state this in the cover letter.<br />

Proofs<br />

Page proofs will be sent to the corresponding author. Please provide an e-mail address to enable page proofs to be sent<br />

as PDF files via e-mail. These should be checked thoroughly for any possible changes or typographic errors. Significant<br />

alterations instigated at this stage by the author will be charged to the author. It is the intention of the Editor to review,<br />

correct and publish your article as quickly as possible. To achieve this it is important that all of your corrections are returned<br />

to us in one all- inclusive mail or fax. Subsequent additional corrections will not be possible, so please ensure that your first<br />

communication is complete.


<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

New Subscription:<br />

Surname:<br />

First Name:<br />

Hospital or Organization:<br />

Date of subscription:<br />

Full mail address:<br />

Subscription Form<br />

The Journal of Tehran University Heart Center<br />

P.O.BOX: Tell: Fax:<br />

E-mail:<br />

Continuation of Subscription:<br />

The annual Subscription and postage rate: 100/000 Rials for Iran and US $ 100 including postage for other countries.<br />

Please liquidate the total amount of Subscription and postal charges into:<br />

Bank: Refah Branch Code: 1232 Account: Tehran Heart Center Account Number: 200001.28<br />

and send the original Bank slip along with Duly completed form of Subscription to the following addre<br />

ss:<br />

Tehran Heart Center,<br />

North Karegar Street,<br />

Tehran, Iran: 1411713138<br />

Tel: +98 21 88029720<br />

Fax: +98 21 88029702<br />

E-mail: jthc@tums.ac.ir<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

New Subscription:<br />

Surname:<br />

First Name:<br />

Hospital or Organization:<br />

Date of subscription:<br />

Full mail address:<br />

Subscription Form<br />

The Journal of Tehran University Heart Center<br />

P.O.BOX: Tell: Fax:<br />

E-mail:<br />

Continuation of Subscription:<br />

The annual Subscription and postage rate: 100/000 Rials for Iran and US $ 100 including postage for other countries.<br />

Please liquidate the total amount of Subscription and postal charges into:<br />

Bank: Refah Branch Code: 1232 Account: Tehran Heart Center Account Number: 200001.28<br />

and send the original Bank slip along with Duly completed form of Subscription to the following addre<br />

ss:<br />

Tehran Heart Center,<br />

North Karegar Street,<br />

Tehran, Iran: 1411713138<br />

Tel: +98 21 88029720<br />

Fax: +98 21 88029702<br />

E-mail: jthc@tums.ac.ir<br />

<strong>TEHRAN</strong> <strong>HEART</strong> <strong>CENTER</strong><br />

The Journal of Tehran University Heart Center

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!