The International Society of Heart and Lung Transplantation Guidelines for the care of heart...

43
SPECIAL FEATURE The International Society of Heart and Lung Transplantation Guidelines for the care of heart transplant recipients Chair: Maria Rosa Costanzo, MD Task Force 1: Chair: Maria Rosa Costanzo, MD; Co-Chairs: Anne Dipchand, MD; Randall Starling, MD Contributing Writers: Allen Anderson, MD; Michael Chan, MD; Shashank Desai, MD; Savitri Fedson, MD; Patrick Fisher, MD; Gonzalo Gonzales-Stawinski, MD; Luigi Martinelli, MD; David McGiffin, MD; Jon Smith, MD Task Force 2: Chair: David Taylor, MD Co-Chairs: Bruno Meiser, MD; Steven Webber, MD Contributing Writers: David Baran, MD; Michael Carboni, MD; Thomas Dengler, MD; David Feldman, MD; Maria Frigerio, MD; Abdallah Kfoury, MD; Daniel Kim, MD; Jon Kobashigawa, MD; Michael Shullo, PhD; Josef Stehlik, MD; Jeffrey Teuteberg, MD; Patricia Uber, PharmD; Andreas Zuckermann, MD Task Force 3: Chair: Sharon Hunt, MD Co-Chair: Michael Burch Contributing Writers: Geetha Bhat, MD; Charles Canter, MD; Richard Chinnock, MD; Marisa Crespo-Leiro, MD; Reynolds Delgado, MD; Fabienne Dobbels, PhD; Kathleen Grady, PhD; Kao W, MD; Jaqueline Lamour, MD; Gareth Parry, MD; Jignesh Patel, MD; Daniela Pini, MD; Jeffrey Towbin, MD; Gene Wolfel, MD Independent Reviewers: Diego Delgado, MD; Howard Eisen, MD; Lee Goldberg, MD; Jeff Hosenpud, MD; Maryl Johnson, MD; Anne Keogh, MD; Clive Lewis, MD; John O’Connell, MD; Joseph Rogers, MD; Heather Ross, MD; Stuart Russell, MD; Johan Vanhaecke, MD INSTITUTIONAL AFFILIATIONS Chair: Costanzo MR: Midwest Heart Foundation, Lombard Illinois, USA Task Force 1: Dipchand A: Hospital for Sick Children, Toronto Ontario, Canada; Starling R: Cleveland Clinic Foundation, Cleveland, Ohio, USA; Anderson A: University of Chicago, Chicago, Illinois, USA; Chan M: University of Alberta, Edmonton, Alberta, Canada; Desai S: Inova Fairfax Hospital, Fairfax, Virginia, USA; Fedson S: University of Chicago, Chicago, Illinois, USA; Fisher P: Ochsner Clinic, New Orleans, Louisiana, USA; Gonzales-Stawinski G: Cleveland Clinic Foundation, Cleveland, Ohio, USA; Martinelli L: Ospedale Niguarda, Milano, Italy; McGiffin D: University of Alabama, Birmingham, Alabama, USA; Smith J: Freeman Hospital, Newcastle upon Tyne, UK Task Force 2: Taylor D: Cleveland Clinic Foundation, Cleveland, Ohio, USA; Meiser B: University of Munich/Grosshaden, Munich, Germany; Baran D: Newark Beth Israel Medical Center, Newark, New Jersey, USA; Carboni M: Duke University Medical Center, Durham, North Carolina, USA; Dengler T: University of Hidelberg, Heidelberg, Germany; Feldman D: Minneapolis Heart Institute, Minneapolis, Minnesota, USA; Frigerio M: Ospedale Niguarda, Milano, Italy; Kfoury A: Intermountain Medical Center, Murray, Utah, USA; Kim D: University of Alberta, Edmonton, Alberta, Canada; Kobashigawa J: Cedar-Sinai Heart Institute, Los Angeles, California, USA; Shullo M: University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Stehlik J: University of Utah, Salt Lake City, Utah, USA; Teuteberg J: University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Uber P: University of Maryland, Baltimore, Maryland, USA; Zuckermann A: University of Vienna, Vienna, Austria. Task Force 3: Hunt S: Stanford University, Palo Alto, California, USA; Burch M: Great Ormond Street Hospital, London, UK; Bhat G: Advocate Christ Medical Center, Oak Lawn, Illinois, USA; Canter C: St. Louis Children Hospital, St. Louis, Missouri, USA; Chinnock R: Loma Linda University Children’s Hospital, Loma Linda, California, USA; Crespo-Leiro M: Hospital Universitario A Coruña, La Coruña, Spain; Delgado R: Texas Heart Institute, Houston, Texas, USA; Dobbels F: Katholieke Universiteit Reprint requests: Amanda W. Rowe, International Society for Heart and Lung Transplantation, 14673 Midway Road, Suite 200, Addison, TX 75001. Telephone: 804-873-2541. Fax: 817-912-1237. E-mail address: [email protected]; [email protected] http://www.jhltonline.org 1053-2498/$ -see front matter © 2010 International Society for Heart and Lung Transplantation. All rights reserved. doi:10.1016/j.healun.2010.05.034

Transcript of The International Society of Heart and Lung Transplantation Guidelines for the care of heart...

S

TGCTCPTCCMJTCCRGIMS

T

http://www.jhltonline.org

1d

PECIAL FEATURE

he International Society of Heart and Lung Transplantationuidelines for the care of heart transplant recipients

hair: Maria Rosa Costanzo, MDask Force 1: Chair: Maria Rosa Costanzo, MD; Co-Chairs: Anne Dipchand, MD; Randall Starling, MDontributing Writers: Allen Anderson, MD; Michael Chan, MD; Shashank Desai, MD; Savitri Fedson, MD;atrick Fisher, MD; Gonzalo Gonzales-Stawinski, MD; Luigi Martinelli, MD; David McGiffin, MD; Jon Smith, MDask Force 2: Chair: David Taylor, MDo-Chairs: Bruno Meiser, MD; Steven Webber, MDontributing Writers: David Baran, MD; Michael Carboni, MD; Thomas Dengler, MD; David Feldman, MD;aria Frigerio, MD; Abdallah Kfoury, MD; Daniel Kim, MD; Jon Kobashigawa, MD; Michael Shullo, PhD;

osef Stehlik, MD; Jeffrey Teuteberg, MD; Patricia Uber, PharmD; Andreas Zuckermann, MDask Force 3: Chair: Sharon Hunt, MDo-Chair: Michael Burchontributing Writers: Geetha Bhat, MD; Charles Canter, MD; Richard Chinnock, MD; Marisa Crespo-Leiro, MD;eynolds Delgado, MD; Fabienne Dobbels, PhD; Kathleen Grady, PhD; Kao W, MD; Jaqueline Lamour, MD;areth Parry, MD; Jignesh Patel, MD; Daniela Pini, MD; Jeffrey Towbin, MD; Gene Wolfel, MD

ndependent Reviewers: Diego Delgado, MD; Howard Eisen, MD; Lee Goldberg, MD; Jeff Hosenpud, MD;aryl Johnson, MD; Anne Keogh, MD; Clive Lewis, MD; John O’Connell, MD; Joseph Rogers, MD; Heather Ross, MD;

tuart Russell, MD; Johan Vanhaecke, MD

INSTITUTIONAL AFFILIATIONSChair: Costanzo MR: Midwest Heart Foundation, Lombard Illinois, USATask Force 1: Dipchand A: Hospital for Sick Children, Toronto Ontario, Canada; Starling R:Cleveland Clinic Foundation, Cleveland, Ohio, USA; Anderson A: University of Chicago, Chicago,Illinois, USA; Chan M: University of Alberta, Edmonton, Alberta, Canada; Desai S: Inova FairfaxHospital, Fairfax, Virginia, USA; Fedson S: University of Chicago, Chicago, Illinois, USA; Fisher P:Ochsner Clinic, New Orleans, Louisiana, USA; Gonzales-Stawinski G: Cleveland Clinic Foundation,Cleveland, Ohio, USA; Martinelli L: Ospedale Niguarda, Milano, Italy; McGiffin D: University ofAlabama, Birmingham, Alabama, USA; Smith J: Freeman Hospital, Newcastle upon Tyne, UKTask Force 2: Taylor D: Cleveland Clinic Foundation, Cleveland, Ohio, USA; Meiser B: Universityof Munich/Grosshaden, Munich, Germany; Baran D: Newark Beth Israel Medical Center, Newark,New Jersey, USA; Carboni M: Duke University Medical Center, Durham, North Carolina, USA;Dengler T: University of Hidelberg, Heidelberg, Germany; Feldman D: Minneapolis Heart Institute,Minneapolis, Minnesota, USA; Frigerio M: Ospedale Niguarda, Milano, Italy; Kfoury A: IntermountainMedical Center, Murray, Utah, USA; Kim D: University of Alberta, Edmonton, Alberta, Canada;Kobashigawa J: Cedar-Sinai Heart Institute, Los Angeles, California, USA; Shullo M: University ofPittsburgh, Pittsburgh, Pennsylvania, USA; Stehlik J: University of Utah, Salt Lake City, Utah, USA;Teuteberg J: University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Uber P: University of Maryland,Baltimore, Maryland, USA; Zuckermann A: University of Vienna, Vienna, Austria.Task Force 3: Hunt S: Stanford University, Palo Alto, California, USA; Burch M: Great Ormond StreetHospital, London, UK; Bhat G: Advocate Christ Medical Center, Oak Lawn, Illinois, USA; Canter C:St. Louis Children Hospital, St. Louis, Missouri, USA; Chinnock R: Loma Linda University Children’sHospital, Loma Linda, California, USA; Crespo-Leiro M: Hospital Universitario A Coruña, La Coruña,Spain; Delgado R: Texas Heart Institute, Houston, Texas, USA; Dobbels F: Katholieke Universiteit

Reprint requests: Amanda W. Rowe, International Society for Heart and Lung Transplantation, 14673 Midway Road, Suite 200, Addison, TX 75001.elephone: 804-873-2541. Fax: 817-912-1237.

E-mail address: [email protected]; [email protected]

053-2498/$ -see front matter © 2010 International Society for Heart and Lung Transplantation. All rights reserved.oi:10.1016/j.healun.2010.05.034

SmgvssilspctoiGht

g

915Costanzo et al. Guidelines for Heart Transplant Care

Leuven, Leuven, Belgium; Grady K: Northwestern University, Chicago, Illlinois, USA; Kao W: Universityof Wisconsin, Madison Wisconsin, USA; Lamour J: Montefiore Medical Center, New York, New York,USA; Parry G: Freeman Hospital, Newcastle upon Tyne, UK; Patel J: Cedar-Sinai Heart Institute, LosAngeles, California, USA; Pini D: Istituto Clinico Humanitas, Rozzano, Italy; Towbin J: CincinnatiChildren’s Hospital, Cincinnati, Ohio, USA; Wolfel G: University of Colorado, Denver, Colorado, USAIndependent Reviewers: Delgado D: University of Toronto, Toronto, Ontario, Canada; Eisen H: DrexlerUniversity College of Medicine, Philadelphia, Pennsylvania, USA; Goldberg L: University of Pennsylvania,Philadelphia, Pennsylvania, USA; Hosenpud J: Mayo Clinic, Jacksonville, Florida, USA; Johnson M:University of Wisconsin, Madison, Wisconsin, USA; Keogh A: St Vincent Hospital, Sidney, New SouthWales, Australia; Lewis C: Papworth Hospital Cambridge, UK; O’Connell J: St. Joseph Hospital, Atlanta,Georgia, USA; Rogers J: Duke University Medical Center, Durham, North Carolina, USA; Ross H:University of Toronto, Toronto, Ontario, Canada; Russell S: Johns Hopkins Hospital, Baltimore, Maryland,USA; Vanhaecke J: University Hospital Gasthuisberg, Leuven, Belgium.J Heart Lung Transplant 2010;29:914–956© 2010 International Society for Heart and Lung Transplantation. All rights reserved.

amp

ricAghrhhctt

TT

Ch

CFt

TI

RC

1

ince the dawn of heart transplantation in the 1960s, theedical care of heart transplant recipients has been

uided by the experience of individual clinicians and hasaried from center to center. Despite many advances inurgical techniques, diagnostic approaches, and immuno-uppressive strategies, survival after heart transplantations limited by the development of cardiac allograft vascu-opathy and by the adverse effects of immunosuppres-ion. The International Society for Heart and Lung Trans-lantation (ISHLT) has made an unprecedentedommitment to convene experts in all areas of heartransplantation to develop practice guidelines for the caref heart transplant recipients. After a vast effort involv-ng 40 writers from 9 countries worldwide, the ISHLTuidelines for the Care of Heart Transplant Recipientsave now been completed and the Executive Summary ofhese guidelines is the subject of this article.

The document results from the work of 3 Task Forceroups:

Task Force 1 addresses the peri-operative care of hearttransplant recipients, including the surgical issues affect-ing early post-operative care; monitoring and treatment ofearly hemodynamic, metabolic, and infectious issues;evaluation and treatment of allosensitization; evaluationand treatment of early coagulopathies; the organization ofa multidisciplinary care team; management of ABO “in-compatible” pediatric heart transplantation; and the use ofextracorporeal membrane oxygenation (ECMO) for thehemodynamic support of pediatric recipients.Task Force 2 discusses the mechanisms, diagnosis, andtreatment of heart transplant rejection; the mechanisms ofaction, dosing, and drug level monitoring of immunosup-pressive drugs as well as their adverse effects and inter-actions with concomitantly used medications; and re-views the major clinical trials and the immunosuppressivestrategies to be used in special clinical situations.Task Force 3 covers the myriad of clinical issues occurringlong-term after heart transplantation, including cardiac allograftvasculopathy, the chronic adverse effects of immunosuppres-sion (neurotoxicity, renal insufficiency, hypertension, bone dis-

ease, diabetes and malignancy), as well as reproductive health,

exercise, psychologic problems, return to work, and operationof motor vehicles after heart transplantation.

It is important to note that each task force was co-chaired bypediatric heart transplant physician who had the specificandate to highlight issues unique to the pediatric heart trans-

lant population and to ensure their adequate representation.As the reader will undoubtedly observe, most of the

ecommendations only achieve a Level of Evidence C,ndicating that these recommendations are based on expertonsensus and not on randomized controlled clinical trials.

concerted effort was also made to highlight the numerousaps in evidence pertaining to many aspects of the care ofeart transplant recipients. This lack of “evidence-based”ecommendations is mostly due to the limited number ofeart transplant recipients worldwide. However, it is theope of all contributing writers and reviewers that the in-reased awareness of the “gaps in evidence” provided byhese guidelines will spur further research in many impor-ant areas of heart transplantation.

ask Force 1: Peri-operative Care of the Heartransplant Recipient

hair: Maria Rosa Costanzo, MD; Co-Chairs: Anne Dipc-and, MD; Randall Starling, MD

Contributing Writers: Allen Anderson, MD; Michaelhan, MD; Shashank Desai, MD; Savitri Fedson, MD; Patrickisher, MD; Gonzalo Gonzales-Stawinski, MD; Luigi Mar-

inelli, MD; David McGiffin, MD; Jon Smith, MD

opic 1: Surgical Issues Impacting Care in themmediate Post-operative Period

ecommendations on Donor Heart Selection:1,2

lass IIa:

. Taking into consideration only the variable of “donor age,”the hearts of donors younger than 45 years will invariablyhave sufficient reserves to withstand the rigors of hearttransplant (HT) even in settings of prolonged ischemictime, recipient comorbidities, and multiple previous recip-

ient operations with hemodynamically destabilizing bleed-

RDC

1

RDC

1

2

3

RCC

1

2

C

1

2

RC

1

RC

1

RC

1

916 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

ing. Hearts from donors between the ages of 45 and 55years should probably be used when the projected ischemictime is � 4 hours and the potential recipient does not havecomorbidities or surgical issues where anything less thanrobust donor heart performance could prove fatal. The useof donor hearts � 55 years should only be used if thesurvival benefit of HT for a recipient unequivocally exceedsthe decrement in early HT survival due to transplantation ofa heart with limited myocardial reserves.

Level of Evidence: B.

ecommendation on the Transplantation of Hearts fromonors with Infection3:lass IIa:

. Hearts from donors with severe infection can be usedprovided that (1) the donor infection is community ac-quired and donor death occurs rapidly (within 96 hours);(2) repeat blood cultures before organ procurement arenegative; (3) pathogen-specific anti-microbial therapy isadministered to the donor; (4) donor myocardial functionis normal; and (5) there is no evidence of endocarditis bydirect inspection of the donor heart. If such hearts areused for transplantation, the recipient should undergosurveillance blood cultures on the first post-operativeday and pathogen-specific anti-biotic therapy should beadministered for an appropriate duration of time.

Level of Evidence: C.

ecommendation on the Transplantation of Hearts fromonors with Potential Drug Toxicities:4–8

lass IIa:

. Hearts from donors with a history of past or current non-intravenous (IV) cocaine abuse can be used for transplantationprovided cardiac function is normal and LVH is absent.

Level of Evidence: C.. In light of current information, the use of hearts from

donors with a history of “alcohol abuse” remains uncer-tain, but is should probably be considered unwise.

Level of Evidence: C.. The use of hearts from donors who have died of carbon

monoxide intoxication can be recommended with cau-tion, although the safety has not been completely estab-lished. It is recommended that these hearts be used pro-vided there is a normal donor electrocardiogram (ECG)and echocardiogram, minimal elevation of cardiac mark-ers, minimal inotropic requirements, a relatively short isch-emic time, a favorable donor to recipient weight ratio and arecipient with normal pulmonary vascular resistance.

Level of Evidence: C.

ecommendations on the Use of Donors with Pre-existingardiac Abnormalities:9,10

lass I:

. As far as the function is concerned, a donor heart shouldnot be used in the presence of intractable ventriculararrhythmias, the need for excessive inotropic support

(dopamine at a dose of 20 �g/kg/min or similar doses of

other adrenergic agents despite aggressive optimizationof pre-load and after-load), discreet wall motion abnor-malities on echocardiography or left ventricular ejectionfraction (LVEF) � 40% despite optimization of hemo-dynamics with inotropic support.

Level of Evidence: B.. A donor heart with a normally functioning bicuspid

aortic valve can be used for HT. Anatomically and he-modynamically abnormal aortic and mitral valves mayundergo bench repair or replacement with subsequenttransplantation of the heart.

Level of Evidence: C.

lass IIa:

. The use of donor hearts with obstructive disease in anymajor coronary artery should be avoided unless the heartis being considered for the alternate list recipients withconcomitant coronary bypass surgery.

Level of Evidence: C.. It would seem appropriate to use hearts from donors with left

ventricular hypertrophy (LVH) provided it is not associatedwith ECG findings of LVH and LV wall thickness is � 14mm.

Level of Evidence: C.

ecommendations on Donor Cardiac Function:lass I:

. As far as the function is concerned, a donor heart should notbe used in the presence of intractable ventricular arrhyth-mias, the need for excessive inotropic support (dopamine ata dose of 20 mcg/kg/min or similar doses of other adren-ergic agents despite aggressive optimization of preload andafter load), discreet wall motion abnormalities on echocar-diography or LV ejection fraction � 40% despite optimi-zation of hemodynamics with inotropic support.

Level of Evidence: B.

ecommendations on Donor-Recipient Size Matching:11,12

lass I:

. As a general rule, the use of hearts from donors whosebody weight is no greater than 30% below that of therecipient is uniformly safe. Furthermore, a male donor ofaverage weight (70 kg) can be safely used for any sizerecipient irrespective of weight. Use of a female donorwhose weight is more than 20% lower than that of a malerecipient should be viewed with caution.

Level of Evidence: C.

ecommendations on Ischemic Times10:lass I:

. As a general rule the ischemic time should be less than4 hours. However, there are situations in which ischemictimes longer than 4 hours are anticipated. Donor heartswith ischemic times longer than 4 hours should only be

accepted when other factors interacting with ischemic

TT

RHC

1

RTC

1

C

1

RP

C

1

2

C

1

Ri(C

1

2

3

4

RVs

917Costanzo et al. Guidelines for Heart Transplant Care

time are ideal, including donor young age, normal car-diac function, and absence of inotropic support.

Level of Evidence: C.

opic 2: Early Post-operative Care of the Heartransplant Recipient

ecommendations on the Post-operative Monitoring ofeart Transplant Recipients:13–31

lass I:

. Peri-operative monitoring of heart transplant recipientsshould include (1) continuous ECG monitoring; (2) post-operative 12-lead ECG; (3) invasive arterial pressuremonitoring; (4) direct measurement of right atrial pres-sure (RAP) or central venous pressure (CVP); (5) mea-surement of left atrial or pulmonary artery wedge pres-sure (PAWP); (6) intermittent measurement of cardiacoutput (CO); (7) continuous measurement of arterialoxygen saturation; (8) intraoperative transesophagealechocardiogram (TEE); (9) continuous assessment ofurinary output.

Level of Evidence: C.

ecommendations on the Management of Peri-operativericuspid Valve Regurgitation:32,33

lass I:

. Tricuspid valve regurgitation identified intraoperativelyand estimated to be moderate or severe (� 2�), shouldbe re-evaluated by transthoracic echocardiogram (TTE)or TEE within 24 hours of HT and closely monitored forthe first few post-operative days. The frequency of sub-sequent follow-up should be guided by clinical and he-modynamic variables.

Level of Evidence: C.

lass II:

. DeVega annuloplasty of the donor tricuspid valve (TV)can be considered to maintain the normal size of the TVannulus.

Level of Evidence: C.

ecommendations on the Management of Peri-operative

Table 1 Properties of Intravenous Vasoactive Drugs Used afte

Peripheralvasoconstriction

Cardiaccontrac

Isoproterenol 0 ����Dobutamine 0 ���Dopamine �� ���Epinephrine ��� ����Milrinone/enoximone 0 ���Norepinephrine ���� ���Phenylephrine ���� 0Vasopressin ���� 0

Adapted and reprinted with permission from Kirklin JK, et al.46

ericardial Effusions:34,35 (

lass I:

. Pericardial effusions occurring after HT should be mon-itored by echocardiogram.

. Percutaneous or surgical drainage should be done when thepericardial effusion causes hemodynamic compromise.

Level of Evidence: C.

lass IIa:

. Pericardial effusions that are not hemodynamically com-promising do not require drainage unless there is a strongsuspicion of an infectious etiology.

Level of Evidence: C.

ecommendations for Peri-operative Vasoactive Drugs Usen Heart Transplant Recipients36–46:See Table 1)lass I:

. Continuous infusion of an inotropic agent should be usedto maintain hemodynamic stability post-operatively. Ino-tropic agents should be weaned as tolerated over the first3 to 5 days. The lowest effective dose should be used.

Level of Evidence: C.. The following therapies are suggested:

a. isoproterenol, 1 to 10 �g/min, orb. dobutamine, 1 to 10 �g/kg/min � dopamine 1 to 10

�g/kg/min, orc. isoproterenol, 1 to 10 �g/min � dopamine 1 to 10

�g/kg/min, ord. milrinone, 0.375 to 0.75 �g/kg/min

Level of Evidence: C.

. Continuous infusion of �-adrenergic agonists includingphenylephrine, norepinephrine, or epinephrine can beused to maintain adequate mean arterial pressure.

Level of Evidence: C.. Low dose vasopressin (0.03–0.1 U/min) or methylene

blue can be added to �-agonist for vasodilatory shock.Level of Evidence: B.

ecommendations for the Medical Management of Rightentricular Dysfunction and Pulmonary Vascular Hyperten-ion After Heart Transplantation36–45,47:

t Transplantation

Peripheralvasodilation

Chronotropiceffect

Arrhythmiarisk

��� ���� ������ � �� � �� �� ���� �� ��0 � �0 0 00 0 0

r Hear

tility

See Figure 1)

C

1

C

1

2

RCC

1

2

3

4

C

1

Fmp

918 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

lass I:

. Inotropic agents that can be used to augment right ven-tricle (RV) function include isoproterenol, milrinone,enoximone, dobutamine, and epinephrine.

Level of Evidence: C.

lass IIa:

. Systemic vasodilators with pulmonary vasodilatingproperties, including nitroglycerine and sodium nitro-prusside, can be used in the absence of systemic hypo-tension.

Level of Evidence: C.. Selective pulmonary vasodilators that can be used in the

management of peri-operative RV dysfunction include(1) prostaglandins (prostaglandin E1 [alprostadil], pros-taglandin I2 [epoprostenol or prostacyclin], inhaled ilo-prost); (2) inhaled nitric oxide; (3) sildenafil.

Level of Evidence: C.

ecommendations on the Peri-operative Use of Mechanicalirculatory Support After Heart Transplantation:48–52

lass I:

. Mechanical circulatory support (MCS) should be initi-

igure 1 Management of right ventricular dysfunction. AV,yocardial infarction; PE, pulmonary embolism; PEEP, positive

ermission from Haddad F, et al.47

ated early if there is failure to wean from cardiopulmo-

nary bypass (CPB) or other evidence of heart allograftfailure such as the requirement for multiple high-doseinotropic agents to permit separation from CPB.

Level of Evidence: B.. MCS should be considered if there is continued or wors-

ening hemodynamic instability, such as decreasing car-diac index (CI) and a falling MVO2 or MVO2 � 50%that is not corrected by appropriate resuscitation.

Level of Evidence: B.. Support for either LV or RV failure should escalate from

pharmacotherapy to IABP to MCS.Level of Evidence: B.

. Small ventricular assist devices (VADs) such as theTandemHeart and Levitronix Centrimag can provide ad-equate support for RV, LV, or biventricular (BiV) fail-ure, and have benefits of ease of implantation, manage-ment, and explant.

Level of Evidence: C.

lass IIa:

. In the presence of hemodynamic instability, cardiac tam-ponade should be excluded by direct surgical explora-tion. The presence of hyperacute/antibody-mediated re-

entricular; CVVH, continuous venovenous hemofiltration; MI,piratory pressure; SR, sinus rhythm. Adapted and reprinted with

atriovend-ex

jection should also be excluded. If hemodynamic

2

C

1

RlC

1

2

C

RCC

1

2

3

4

5

6

7

C

1

2

RF6

C

1

2

3

4

5

6

919Costanzo et al. Guidelines for Heart Transplant Care

instability persists in the absence of cardiac tamponade,MCS should be considered.

Level of Evidence: C.. The timing MCS discontinuation should be guided by

evidence of graft recovery. If there is no evidence ofgraft functional recovery within 3 to 4 days, hyperacuteand antibody-mediated rejection should be excluded andthe option of listing for repeat HT may be considered.

Level of Evidence: C.

lass IIb:

. Use of ECMO support in adults requires consideration ofthe risk of infection, immobility, and need for anti-coagulation.

Level of Evidence: C.

ecommendations for the Management of Early Heart Al-ograft Dysfunction in Pediatric Recipients:53–60

lass IIb:

. The increased risk of post-operative RV dysfunctionmust be carefully evaluated in children, although evi-dence suggests that children can safely undergo HT de-spite elevation of pulmonary vascular resistance (PVR)above values considered unsafe in adults.

Level of Evidence: C.. Contrary to the experience and practice in adults, the first

choice for support in the setting of primary graft failure(PGF) in the pediatric setting should be ECMO.

lass IIa, Level of Evidence C.

ecommendations for the Peri-operative Management ofardiac Arrhythmias in Heart Transplant Recipients:61–67

lass I:

. Pharmacologic chronotropic agents, including isoproter-enol and theophylline can be used in the peri-operativesetting to increase heart rate.

Level of Evidence: B.. Atrial and ventricular temporary epicardial pacing wires

should be placed at the time of HT even if the initialrhythm is sinus.

Level of Evidence: B.. After HT, temporary pacing should be initiated in the

setting of relative bradycardia to maintain heart rates of� 90 beats/min.

Level of Evidence: B.. Pacing guidelines of the American College of Cardiol-

ogy (ACC)/American Heart Association (AHA)/HeartRhythm Society (HRS) and the European Society ofCardiology (ESC) lack recommendations specific fortemporary pacing early after HT. Recommendations forpermanent pacing exist for inappropriate chronotropicresponse 3 weeks after HT. Standard atrium-paced, atri-um-sensed, inhibited-rate modulation (AAIR) or dual-paced, dual-sensed, dual-response to sensing, rate mod-ulation (DDDR) pacemakers are preferable.

Level of Evidence: C.

. Treatment of tachyarrhythmias should be aimed at ratecontrol.

Level of Evidence: B.. Persistent tachyarrhythmias, whether atrial or ventricu-

lar, should prompt investigation of possible rejection andelectrophysiological evaluation if rejection is absent.

Level of Evidence: B.. Sustained ventricular tachycardia (SVT) should be eval-

uated with both an angiogram and an endomyocardialbiopsy (EMB).

Level of Evidence: B.

lass IIa:

. The Class III anti-arrhythmics sotalol and amiodaronecan be safely used in HT recipients and have minimalinteraction with immunosuppressive agents.

Level of Evidence: C.. Non-dihydropyridine calcium channel blockers (CCBs)

and �-blockers may be used in HT recipients for ratecontrol.

Level of Evidence: B.

ecommendations for Peri-operative Renal Function andluid Status Management in Heart Transplant Recipients:8–71

lass I:

. The CVP should be maintained between 5 and 12 mmHg, a level that provides adequate cardiac filling pres-sures without causing RV overload.

Level of Evidence: C.. Colloid replacement is generally preferred in the first 24

hours after HT; blood, if indicated, is the first choice.Level Evidence: C.

. Compatible blood products may be safely administeredafter HT without increasing the risk for rejection. In thesetting of ABO incompatible pediatric HT special caremust be taken in the selection of compatible products toaccount for both donor and recipient blood types.

Level of Evidence: B.. Blood products should be leukocyte-depleted. Blood

products should be cytomegalovirus (CMV) negative ifdonor and recipient are CMV negative.

Level of Evidence: B.. IV loop diuretics are used to decrease volume overload.

In addition to intermittent IV bolus, continuous IV infu-sion of loop diuretics with or without sequential nephro-nal blockade using thiazide diuretics or aldosterone an-tagonists may be necessary.

Level of Evidence: C.. Hemodialysis for renal failure should be initiated early

for both volume management and renal replacement. Ifthe recipient is anuric, oliguric, or has a sharp rise in sCrwithin 2 to 4 hours after HT, then hemodialysis may benecessary.

Level of Evidence: B.

C

1

C

1

RHC

1

C

1

2

RmC

1

2

3

4

Ri(C

1

2

3

RlC

1

RTC

1

920 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

lass IIa:

. Ultrafiltration should be considered if RAP remains ele-vated (� 20 mm Hg) despite pharmacologic interventions.

Level of Evidence: B.

lass IIb:

. Delay of initiation of calcineurin inhibitor (CNI) therapyshould be considered if there is significant pre-operativerenal insufficiency or deterioration of kidney function inthe first 2 post-operative days.

Level of Evidence: C.

ecommendations for the Peri-operative Management ofyperglycemia in Heart Transplant Recipient:72,73

lass I:

. Oral hypoglycemic agents should be discontinued pre-operatively.

Level Evidence: C.

lass IIa:

. A continuous infusion insulin regimen should be used tomaintain blood glucose below 200 mg/dL during theintensive care unit (ICU) stay.

Level of Evidence: B.. Aggressive management of hyperglycemia should be

continued for the duration of hospitalization.Level of Evidence: C.

ecommendations for Anti-bacterial Prophylaxis/Treat-ent74:

lass I:

. Pre-operative anti-biotic prophylaxis should be used be-fore the transplant operation.

Level of Evidence: B.. Drugs should be selected based upon their activity

against usual skin flora, specifically Staphylococcus spe-cies.

Level of Evidence: B.. If a chronically infected device such as a VAD or a

pacemaker is present, then peri-operative anti-bioticsshould be selected based on microbiologic sensitivities.

Level of Evidence: B.. In the event that the donor had an ongoing bacterial

infection, a course of suitable anti-biotics should beconsidered.

Level of Evidence: B.

ecommendations for Peri-operative Anti-viral Prophylaxisn Heart Transplant Recipients75:See Table 2)lass I:

. Prophylaxis against CMV should be initiated within 24to 48 hours after HT.

Level of Evidence: A.. The CMV serologic status of the donor and recipient

may be used to stratify the patient as low-risk, interme-

diate-risk, or high-risk for developing a CMV infection.

Level of Evidence: A.. Intravenous ganciclovir may be administered to interme-

diate and high-risk patients, whereas patients at low-riskfor CMV infection may only receive anti-herpes simplexvirus prophylaxis with acyclovir. (See Table 3.)

Level of Evidence: A.

ecommendations for Peri-operative Anti-Fungal Prophy-axis in Heart Transplant Recipients76:lass I:

. Anti-fungal prophylaxis to prevent mucocutaneous can-didiasis should be initiated once the recipient is extu-bated. The agents most commonly used are nystatin(4–6 mL [400,000 to 600,000 units] 4 times daily, swishand swallow) or clotrimazole lozenges (10 mg).

Level of Evidence: C.

ecommendations for Anti-Protozoal Prophylaxis in Heartransplant Recipients77:lass I:

. Prophylaxis against Pneumocystis jiroveci (formerlyPneumocystis carinii) pneumonia and Toxoplasma gon-dii (in indicated cases) should also be initiated in theearly post-operative period. Trimethoprim/sulfamethox-azole (80 mg TMP/160 mg SMZ, 1 single- or double-strength tablet per day) is the most commonly usedmedication. In the setting of a sulfa allergy or glucose-6-phosphate dehydrogenase deficiency, alternative regi-mens can be used, including: (1) Aerosolized pentami-dine (AP) isethionate (300 mg every 3–4 weeks). (2)Dapsone (diaminodiphenylsulfone) with or without TMPor pyrimethamine (50–100 mg/day). Pyrimethaminemay be administered weekly (25 or 50 mg) to supple-

Table 2 Typical Recommendations for the Prevention ofCytomegalovirus in Heart Transplant Recipients

Group Recommendations/Options

D�/R– Oral ganciclovir (1000 g PO TID) or valganciclovir(900 mg PO/day) for 3 months

orIV ganciclovir (5–10 mg/kg/day) for 1–3 monthsPreemptive therapy generally not preferred due

to high risk of diseaseSome HT centers will add CMV immune globulin

for high risk patientsR� Oral ganciclovir (1000 g PO TID) or valganciclovir

(900 mg PO/day) for 3 monthsorIV ganciclovir (5–10 mg/kg/day) for 1–3 monthsorPreemptive therapy. Monitor with nucleic acid

testing or CMV antigenemia assayTherapy with IV ganciclovir or oral valganciclovir

CMV, cytomegalovirus; D, donor; HT, heart transplant; IV, intrave-nous; PO, oral (per os); R, recipient; TID, 3 times daily.

ment dapsone (50–100 mg/day). Dapsone is metabolized

RaC

1

2

TARA

RHC

1

2

3

4

5

Rld(C

1

921Costanzo et al. Guidelines for Heart Transplant Care

via the hepatic cytochrome P-450 system (CYP3A). (3)Atovaquone (1500 mg PO QD). (4) Clindamycin andpyrimethamine.

Level of Evidence: B.

ecommendations for Peri-operative Infection Prophylaxisnd Treatment in Pediatric Heart Transplant Recipients:74–77

lass IIb:

. IV anti-fungal prophylaxis should be considered for in-fants (� 1 year of age) with an open chest and/or requir-ing ECMO support in the peri-operative period.

Level of Evidence: C.. Prophylaxis for Pneumocystis jiroveci should be insti-

tuted for a minimum of 3 months up to a maximum of 24months after HT.

Level of Evidence: C.

opic 3: Evaluation of Allosensitization,pproaches to Sensitized Heart Transplantecipients, and Hyperacute and Delayedntibody-Mediated Rejection

ecommendations for the Evaluation of Donor/Recipientistocompatibility:78–84

lass I:

. Screening panel reactive antibodies (PRA) should beperformed in all HT candidates. When the PRA is ele-vated (�10%) further evaluation is recommended.

Level of Evidence: C.. The specificity of circulating antibodies should be deter-

Table 3 Examples of Desensitization Therapies

Therapy Dose

Plasmapheresis (A, F) 1.5 volume exchanges

Intravenous immunoglobulin(IV Ig)

(A, B) 2g/kg IV divided over 2

(C) 2–3 g/kg IV divided over 4(D) 0.1 mg/kg IV(E) 100 mg/kg IV(F) 20 g (of 10% IV Ig)(G) 150 g (of 10% IV Ig) divid

Rituximab (A) 1 g IV(C, E) 375 mg/m2

(G) 500 mg

Cyclophosphamide(used in the past)

(A) 1 mg/kg orally

(C) 0.5 �g/m2

(D) 1 mg/kg orally

(A) UCLA; (B) Stanford University; (C) University of Maryland; (D) Un(G) University of Berlin.

Adapted from Kobashigawa J, et al.80

mined with a solid-phase assay such as flow-cytometry,

if possible, in a regional certified human leukocyte an-tigen (HLA) laboratory.

Level of Evidence: C.. The complement fixation capability of detected antibod-

ies should be reported.Level of Evidence: C.

. The anti-HLA class I and II specificities (ie, any HLAantibody directed against HLA-A, HLA-B, HLA-Cw,HLA-DR, and HLA-DQ antigens) should be defined. Inthe absence of international standards, each transplantcenter must define the threshold of antibody levels usedto define which specific donor HLA antigens confer anunacceptable rejection risk.

Level of Evidence: C.. The virtual crossmatch, which compares recipient anti-

HLA antibody specificities with donor HLA antigens,should be routinely used to increase the donor pool forsensitized recipients.

Level of Evidence: C.

ecommendations for the Risk-Assessment and Prophy-axis Strategies for Allosensitized Heart Transplant Candi-ates80,85:See Table 3)lass IIa:

. A complete patient sensitization history, including pre-vious PRA determinations, blood transfusions, pregnan-cies, implant of homograft materials, previous transplan-tation, and use of a VAD is required to assess the risk ofheart allograft anti-body-mediated rejection.

Frequency

(A) 5 consecutive days(B) 5 times, every other day(C) 2–3 times/week until transplant(D) 5 times, every other day, every 2–4 weeks(A) Every 2–4 weeks

(D) Every 2–4 weeks(E) Every 4 weeks

er 3 rounds (G) Every 4 weeks(A) Weekly � 4(C) �2 doses(E) Weekly � 4(G) Every 2 weeks(A) Daily

of Toronto; (E) University of Wisconsin; (F) Loyola University Chicago;

days

days

ed ov

iversity

Level of Evidence: C.

2

3

C

1

2

3

Rt(C

1

2

3

4

5

6

7

RRC

1

2

3

RPC

1

T

922 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

. A PRA � 10% indicates significant allosensitization and itshould raise the question of whether therapies aimed atreducing allosensitization should be instituted to minimizethe need for a prospective donor/recipient crossmatch.

Level of Evidence: C.. The results of the retrospective donor recipient cross-

match may be considered to make decisions regardingimmunosuppressive therapy.

Level of Evidence: C.

lass IIb:

. Desensitization therapy should be considered when thecalculated PRA is considered by the individual trans-plant center to be high enough to significantly decreasethe likelihood for a compatible donor match or to de-crease the likelihood of donor heart rejection where un-avoidable mismatches occur.

Level of Evidence: C.. Choices to consider as desensitization therapies include

IV immunoglobulin (Ig) infusion, plasmapheresis, eitheralone or combined, rituximab, and in very selected cases,splenectomy.

Level of Evidence: C.. A large randomized controlled clinical trial is needed to

assess the effectiveness of desensitization strategies andtheir impact on outcomes after HT.

Level of Evidence: C.

ecommendations for Monitoring of Allosensitization Sta-us of Heart Transplant Candidates and Recipients78,80,85:Table 4)lass IIb:

. The presence of anti-HLA antibodies should be regularlymonitored in allosensitized patients undergoing desensi-tizing therapies until a compatible heart allograft be-comes available.

Level of Evidence: C.. In ambulatory, non-sensitized HT candidates it is rea-

sonable to measure anti-HLA antibodies every 6 months.Level of Evidence: C.

. In HT candidates requiring blood transfusions, anti-HLAantibodies determination should be repeated 2 to 4 weekslater and prospective donor/recipient crossmatch is re-quired in the interim period if a suitable donor organbecomes available.

Table 4 Panel-Reactive Antibody Screening Frequency After O

PRA

Number of heart tra

1 mon 2 mon 3 mon 4–6 mo

Negative 10 2 8 16Positive 33 8 6 2

PRA, panel reactive antibody; SE, sensitizing events.Adapted from Betkowski AS, et al.78

Level of Evidence: C. H

. No uniform recommendations exist as to the frequencyof anti-HLA antibody determinations after an infectionor during MCS.

Level of Evidence: C.. Circulating immunoglobulins should be measured before

and after plasmapheresis or immunoabsorption.Level of Evidence: C.

. Lymphocyte sub-populations should be measured beforeand after the use of rituximab.

Level of Evidence: C.. In addition to the post-operative retrospective cross-

match, donor-specific antibodies levels should be ob-tained when antibody-mediated rejection (AMR) is sus-pected or confirmed by EMB.

Level of Evidence: C.

ecommendations for the Treatment of Antibody-Mediatedejection:80,86

lass IIa:

. Initial therapy of AMR can include immunoadsorptionand corticosteroid (CS) or plasmapheresis/low dose ofIV Ig and CS.

Level of Evidence: C.. Rituximab can be added to reduce the risk of recurrent

rejection.Level of Evidence: C.

. Changes in therapy, which can be considered for mainte-nance immunosuppression in patients who experienceAMR, can include switch to tacrolimus (TAC) in patientsreceiving cyclosporine (CYA)-based immunosuppression,increased doses of mycophenolate mofetil (MMF), and CS.

Level of Evidence: C.

ecommendations for the Approach to Allosensitization inediatric Heart Transplant Recipients:76,77,87

lass IIb:

. The HT can be carried out in highly sensitized pediatricpatients without a prospective crossmatch or virtualcrossmatch at centers experienced in pediatric HT acrossa positive crossmatch.

Level of Evidence: C.

opic 4: Management of ABO “Incompatible”

Assessment

t centers screening at each interval

1 year Variable SE Other Total

7 4 16 2 65. . . . . . . . . . . . 65

riginal

nsplan

n

eart Transplant Recipients

R“C

1

2

3

RpC

1

2

RtRC

1

Ri(C

1

2

3

RpC

1

2

RpC

1

923Costanzo et al. Guidelines for Heart Transplant Care

ecommendations for the Selection of Candidates for ABOIncompatible” Heart Transplant:88,89

lass IIa:

. The upper limit of age or isohemagglutinin titer forABO-incompatible pediatric HT remains unclear.

Level of Evidence: C.. ABO-incompatible HT can be safely performed in the

pediatric population in the presence of positive isohem-agglutinin titers against the donor organ.

Level of Evidence: C.. ABO-incompatible HT, especially in the presence of

donor-specific isohemagglutinins � 1:4, should be per-formed in an experienced center.

Level of Evidence: C.

ecommendation for the Intraoperative Care of ABO “Incom-atible” Heart Transplant Recipients:88,89

lass IIa:

. ABO-incompatible HT can be undertaken by performingplasma exchange using the CPB circuit to remove donorspecific isohemagglutinins.

Level of Evidence: C.. Plasma exchange using the CPB circuit allows the safe

transplantation of ABO-incompatible organs without theneed of aggressive pre-operative immunosuppressivetherapies or splenectomy.

Level of Evidence: C.

ecommendations for the Monitoring of Isohemagglu-inin Levels in ABO “Incompatible” Heart Transplantecipients:88,90

lass IIa:

. Serial measurements of isohemagglutinin titers shouldbe done in the post-operative period. Decisions aboutwhether immunosuppressive therapy must be modifiedshould be based not only on the change in isohemagglu-tinin titers but also on clinical or pathologic evidence ofrejection.

Table 5 Match of Blood Products to Specific ABO-Incompatib

Blood groupRed blood cells(plasma depleted)

FreplaRecipient’s Donor’s

O A O AO B O BO AB O ABA B A ABA AB A ABB A B ABB AB B AB

aSecond choice.

Level of Evidence: C.

ecommendations for the Administration of Blood Productsn ABO “Incompatible” Heart Transplant Recipients88–90:See Table 5)lass IIa:

. Whole blood products should never be administered to achild who has received an ABO-incompatible HT, andthe families should be educated to communicate this factto other caregivers in the case of any future medicalemergency or surgery. Group O red blood cells andgroup AB blood elements are safe for every blood groupcombination.

Level of Evidence: C.. If red blood cells transfusions are given to any ABO-

incompatible HT recipient, red blood cell units should bematched based on the HT recipient’s ABO blood type.

Level of Evidence: C.. If platelets and/or plasma preparations are needed in

ABO-incompatible HT recipients, these blood productsshould be matched based on the donor’s ABO bloodtype.

Level of Evidence: C.

ecommendations for Immunosuppression in ABO “Incom-atible” Heart Transplant Recipients:88,89,91

lass IIa:

. Standard (triple) immunosuppression with a CNI, ananti-proliferative agent, and CS can be used in childrenundergoing ABO-incompatible HT without an increasedrisk of rejection.

Level of Evidence: B.. Immunosuppression management beyond the peri-oper-

ative period is similar to that of the ABO-compatiblepediatric HT population.

Level of Evidence: B.

ecommendation for Rejection Surveillance in ABO “Incom-atible” Heart Transplant Recipients:88–90

lass IIa:

. Rejection surveillance in ABO-incompatible HT recipients

rt Transplant Scenario

enCryoprecipitate

Platelets (managed similarlyto plasma)

2nd choice

A A O concentrateB B O concentrateAB, A or B AB A or B concentrateAB, or Ba AB B concentrateAB, A or Ba AB A or B concentrateAB, or Aa AB A concentrateAB, A or Ba AB A or B concentrate

le Hea

sh frozsma

is the same as that of the ABO-compatible HT population.

TS

RHC

1

2

3

C

1

2

3

4

RfC

1

2

3

RRC

1

2

3

4

5

opqVtbndcHo

RCC

1

C

1

924 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

Level of Evidence: C.

opic 5: Coagulopathies in Heart Transplanturgery

ecommendations for the Evaluation of Hemostasis ineart Transplant Recipients92:lass I:

. A history of bleeding (including details of family history,previous excessive post-traumatic or post-surgical bleed-ing) and of the use of any medications that alter coagu-lation should be obtained from the patient.

Level of Evidence: C.. Screening coagulation tests of prothrombin time (PT),

activated partial thromboplastin time (aPTT), and plate-lets counts should be measured immediately before HTsurgery.

Level of Evidence: C.. The activated clotting time (ACT) should be obtained at

multiple points during the HT surgery to gauge theactivity of heparin during each phase of the HT surgery.

Level of Evidence: C.

lass IIa:

. Thromboelastography may be useful during the HT surgeryto further elucidate the status of the patient’s hemostasis.

Level of Evidence: C.. Platelet function can be measured either by platelet aggre-

gometry or by a point of care assay such as the plateletsfunction assay 100 (PFA-100) during the HT surgery.

Level of Evidence: C.. Fibrinogen levels and D-Dimer values should be measured

post-operatively because these are tests of fibrinolysis andcorrelate with the risk of bleeding after HT surgery.

Level of Evidence: C.. Thromboelastography may be repeated after HT surgery

to monitor patients’ hemostasis.Level of Evidence: C.

ecommendations for the Reversal of Anti-coagulation be-ore Heart Transplantation:93–96

lass I:

. Pre-operatively, the international normalized ratio (INR)should be reduced to � 1.5.

Level of Evidence: C.. Low doses of vitamin K (2.5–5.0 mg) given IV are

preferable to high doses because they are associated witha lower risk of anaphylaxis.

Level of Evidence: C.. Given the need for rapid normalization of the INR, chron-

ically anti-coagulated patients about to undergo HT shouldreceive vitamin K in conjunction with fresh frozen plasma(FFP), prothrombin plasma concentrates (PCCs), or recom-binant factor VII (rFVII), depending on their availabilityand the patient’s renal and hepatic functions.

Level of Evidence: C.

ecommendations for Anti-coagulation in Heart Transplantecipients:97–99

lass IIa:

. The absence of platelet factor 4/heparin antibodiesshould be confirmed.

Level of Evidence: C.. The use of unfractionated heparin should be restricted to

the operative procedure itself. Low-molecular-weightheparin is not recommended, due to a longer half-lifethan unfractionated heparin and the inability to fullyreverse its effect with protamine.

Level of Evidence: C.. Alternative anti-coagulants can be used pre-operatively

and post-operatively in patients with history of heparin-induced thrombocytopenia (HIT) in whom the plateletcount has recovered but immunoglobulin G (IgG) anti-bodies to the platelet factor 4/heparin complex are stillpresent.

Level of Evidence: C.. Patients with abnormal hepatic and normal renal func-

tion can be treated with lepirudin, danaparoid, orfondaparinux, whereas those with abnormal renal andnormal hepatic function can receive argatroban at stan-dard doses or lepirudin at reduced doses.

Level of Evidence: C.. Patients with both renal and hepatic dysfunction can be

treated with argatroban or bivalirudin at reduced doses.Level of Evidence: C.

Gaps in Evidence:Transfusion strategies are not well studied. Consensus

pinion drives the decision of when to transfuse bloodroducts. Expert opinions on which clinical situations re-uire transfusions are highly variable. Recombinant factorIIa has not been tested in controlled clinical trials and

herefore there is little evidence to support its use in aleeding cardiac surgery patient. Tranexamic acid and ami-ocaproic acid have not been evaluated in a definitive ran-omized study. Very few studies have been performed spe-ifically in HT recipients. Thus, the recommendations forT are extrapolated from evidence regarding achievementf hemostasis in general cardiac surgery.

ecommendations for the Pharmacologic Management ofoagulopathies in Heart Transplant Recipients:100–105

lass I:

. Transfusion of coagulation factors is necessary for ade-quate hemostasis. Thus, fresh frozen plasma and plate-lets should be transfused based on measured levels. Fi-brinogen infusion for massive bleeding and inadequatefibrinogen levels is needed to control blood loss.

Level of Evidence: C.

lass IIa:

. Tranexamic acid and epsilon-aminocaproic acid bothhave anti-fibrinolytic activity and can be used beforeCPB to reduce the risk of bleeding in selected patients.

Level of Evidence: B.

C

1

C

1

2

Tt

RtC

1

2

C

1

2

C

1

2

TOF

RMc(C

1

2

RdC

1

2

REC

1

2

925Costanzo et al. Guidelines for Heart Transplant Care

lass IIb:

. Recombinant factor VIIa may be used in cases of intrac-table or excessive bleeding with HT surgery.

Level of Evidence: C.

lass III:

. Although aprotinin can reduce bleeding during HT sur-gery, its routine use is not recommended due to anincreased risk of adverse clinical events.

Level of Evidence: B.. Desmopressin is not recommended for routine use be-

cause its modest reduction in bleeding has been associ-ated with adverse clinical events.

Level of Evidence: A.

opic 6: Documentation and Communication withhe Multidisciplinary Team

ecommendations for the Documentation and Communica-ion with the Multidisciplinary Team:106–108

lass I:

. Transplant centers must have a multidisciplinary ap-proach to patient management.

Level of Evidence: C.. The HT team should have regularly scheduled meetings

of all disciplines involved.Level of Evidence: C.

lass IIa:

. Social work and psychiatry specialists should be inte-grated into the patient management team.

Level of Evidence: B.. Transplant centers should strive to have specialty-trained

pharmacists or physicians with expertise in pharmacol-ogy as part of the multidisciplinary team.

Level of Evidence: B.

lass IIb:

. Integration of input from pharmacists and infectious dis-ease specialists is important during the development oftreatment protocols for HT recipients.

Level of Evidence: B.. Dieticians should be involved in the care of HT recipi-

ents to provide input regarding prevention of weight gainand maintenance of glucose control.

Level of Evidence: C.

opic 7: Use of Extracorporeal Membranexygenation for the Management of Primary Graftailure in Pediatric Heart Transplant Recipients

ecommendations on the Indications for Extracorporealembrane Oxygenation in Pediatric Heart Transplant Re-ipients109–114:See Table 6)

lass IIa: C

. The use of ECMO should be considered when there isfailure to separate from CPB after all correctable causesof such failure have been excluded.

Level of Evidence: C.. ECMO should be promptly instituted when progressive

heart allograft dysfunction occurs post-operatively.Level of Evidence: C.

ecommendations for the Conduct of ECMO Support in Pe-iatric Heart Transplant Recipients:115,116

lass IIa:

. The amount of circulatory support provided by ECMOshould be sufficient to achieve adequate systemic perfu-sion and oxygen delivery while waiting for the myocar-dium to recover.

Level of Evidence: C.. Left heart distension during ECMO support should be

aggressively treated because it will compromise pulmo-nary function and impede LV recovery.

Level of Evidence: C.

ecommendations for the Timing of Discontinuation ofCMO Support in the Setting of Primary Graft Failure117:lass IIa:

. Clinical and echocardiographic variables should be seri-ally assessed to determine if myocardial recovery isoccurring.

Level of Evidence: C.. Objective signs of recovery should lead to weaning and

discontinuation of ECMO support.Level of Evidence: C.

Table 6 Potential Causes of Primary Graft Failure AfterPediatric Heart Transplantation

Donor issues● Poor donor organ preservation● Poor donor quality

● Diminished echocardiographic ejection fraction● Requirement for high inotropic support● Elevated blood troponin I level

● Prolonged ischemic time● Large donor (donor-to-recipient weight ratio � 2.0)● Small donor (donor-to-recipient weight ratio � 1.0)● Prolonged donor cardiopulmonary resuscitation times● Anoxia as cause of death● Non-identical blood type● Donor ageRecipient issues● Pre-transplantation diagnosis of congenital heart disease● Previous sternotomy● Elevated pulmonary vascular resistance● Pre-transplantation need for extracorporeal membrane

oxygenator● Pre-transplantation need for ventilatory support

Adapted from Huddleston CB, et al.114

lass IIb:

1

1

2

3

4

5

6

T

CS

bMMSP

T

RcC

1

2

3

4

C

4

RAC

1

2

C

1

C

1

2

3

4

5

6

T

926 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

. Lack of objective evidence of myocardial recoverywithin 3 to 5 days should prompt consideration of eitherinstitution of long term MCS as a bridge to recovery orHT or withdrawal of life-sustaining therapy.

Level of Evidence: C.

Gaps in Evidence:

. The optimal modality for surveillance of adverse neuro-logic events during ECMO support for PGF is unknown.

. Optimal infection prophylaxis in the immunosuppressedpatient receiving ECMO support for PGF is unknown.

. Optimal renal-sparing immunosuppression protocol(s) inpatients receiving ECMO support for PGF is unknown.

. The duration of time waiting for recovery of myocardialfunction in the setting of PGF beyond which recovery isunlikely is unknown.

. The role of more intermediate and long-term MCS inpatients with myocardial recovery insufficient to allowseparation from ECMO within 5 to 7 days is unknown.

. Risk factors for poor outcomes after retransplantation inECMO-supported HT recipients are unknown.

ask Force 2: Immunosuppression and Rejection

hair: David Taylor, MD; Co-Chairs: Bruno Meiser, MD;teven Webber, MD

Contributing Writers: David Baran, MD; Michael Car-oni, MD; Thomas Dengler, MD; David Feldman, MD;aria Frigerio, MD; Abdallah Kfoury, MD; Daniel Kim,D; Jon Kobashigawa, MD; Michael Shullo, PhD; Josef

tehlik, MD; Jeffrey Teuteberg, MD; Patricia Uber,harmD; Andreas Zuckermann, MD

opic 1: Rejection Surveillance

ecommendations for Rejection Surveillance by Endomyo-ardial Biopsy in Heart Transplant Recipients:118–120

lass IIa:

. It is reasonable to utilize EMB in a HT candidate sus-pected of having an infiltrative cardiomyopathy or aninflammatory process, such as giant cell myocarditis,amyloidosis, or sarcoidosis.

Level of Evidence: C.. The standard of care for adult HT recipients is to perform

periodic EMB during the first 6 to 12 post-operativemonths for surveillance of HT rejection.

Level of Evidence: C.. The standard of care in adolescents should be similar to

that in adults, including surveillance EMB for heart al-lograft rejection for 6 to 12 months after HT. In youngerchildren, especially infants, it is reasonable to utilizeechocardiography as a screening tool to reduce the fre-quency of EMB.

Level of Evidence: C.. After the first post-operative year, EMB surveillance for

an extended period of time (eg, every 4–6 months) isrecommended in HT recipients at higher risk for late

acute rejection, to reduce the risk for rejection with L

hemodynamic compromise, and to reduce the risk ofdeath in African-American recipients.

Level of Evidence: C.

lass IIb:

. The use of routine EMB later than 5 years after HT isoptional in both adults and children, depending on clin-ical judgment and the risk for late allograft rejection.

Level of Evidence: C.

ecommendations for the Non-Invasive Monitoring ofcute Heart Transplant Rejection:120–135

lass IIa:

. In centers with proven expertise in ventricular evokedpotentials (VER) monitoring, intramyocardial electro-grams recorded non-invasively with telemetric pacemak-ers can be used for rejection surveillance in patients atlow risk for rejection.

Level of Evidence: C.. Gene Expression Profiling (Allomap) can be used to rule

out the presence of ACR of grade 2R or greater inappropriate low-risk patients, between 6 months and 5years after HT.

Level of Evidence: B.

lass IIb:

. Use of echocardiography as primary monitoring modal-ity for acute heart allograft rejection in infants can beconsidered as an alternative to surveillance EMB.

Level of Evidence: C.

lass III:

. The routine clinical use of electrocardiographic param-eters for acute heart allograft rejection monitoring is notrecommended.

Level of Evidence: C.. The use of echocardiography as an alternative to EMB

for rejection monitoring is not recommended.Level of Evidence: C.

. The routine clinical use of MRI for acute allograft rejec-tion monitoring is not recommended.

Level of Evidence: C.. The use of brain natriuretic peptide (BNP), troponin I or

T, or C-reactive protein (CRP) levels for acute heartallograft rejection monitoring is not recommended.

Level of Evidence: C.. The use of systemic inflammatory markers for acute

heart allograft rejection monitoring is not recommended.Level of Evidence: C.

. Routine use of non-invasive testing modalities (ECG,imaging, or biomarkers) is not recommended as the pri-mary method for acute heart allograft rejection surveil-lance in older children and adolescents.

Level of Evidence: C.

opic 2: Monitoring of Immunosuppressive Drug

evels

Rs(C

1

C

1

2

3

4

5

6

927Costanzo et al. Guidelines for Heart Transplant Care

ecommendations for the Monitoring of Immunosuppres-ive Drug Levels136–149:See Table 7)lass I:

. The use of the microemulsion formulation of CYA isrecommended because it is associated with more favor-able pharmacokinetic features compared with the oil-

Table 7 Drugs That Affect the Levels of Tacrolimus,Cyclosporine, Sirolimus, and Everolimus

Decrease immunosuppressionlevels

Increase immunosuppressionlevels

Anti-epileptics Anti-microbialsCarbamazepine ClarithromycinFosphenytoin ErythromycinPhenobarbital Metronidazole and tinidazolePhenytoin Quinupristin/dalfopristin

LevofloxacinAnti-microbials Anti-fungalsCaspofungin ClotrimazoleNafcillin ItraconazoleRifabutin KetoconazoleRifampin FluconazoleRifapentine Posaconazole

VoriconazoleAnti-retroviral Therapy Anti-retroviral therapyEfavirenz Protease inhibitors (general)Etravirine AmprenavirNevirapine Atazanavir

DarunavirFosamprenavirIndinavirNelfinavirRitonavirSaquinavirTipranavir

Others CardiovascularAntacids containing

magnesium, calcium, oraluminum (tacrolimus only)

Amiodarone

Deferasirox DiltiazemModafinil VerapamilSt. John’s wortThalidomideTiclopidineTroglitazone

NutraceuticalsBitter orangeGrapefruit juiceOthersRilonaceptTheophyllineCimetidineFluvoxamineGlipizideGlyburideImatinibNefazodone

based compound.

Level of Evidence: B.

lass IIa:

. At present, 2-hour post-dose (C2) levels should not re-place 12-hour trough (C0) concentrations for routinemonitoring of CYA exposure in most patients, but maybe useful in selected patients in whom a better charac-terization of the pharmacokinetic profile of CYA is de-sired.

Level of Evidence: B.. Measurement of 12-hour trough CYA concentration is

the recommended form of therapeutic drug monitoringfor routine clinical use. The target levels are dependenton the method used (high-performance liquid chroma-tography [HPLC] vs enzyme multiplied immunoassaytechnique [EMIT] vs cloned enzyme donor immunoas-say method [CEDIA]), concomitant immunosuppres-sion, toxicity risks, and time after HT. In general, whenused in conjunction with azathioprine (AZA) or a my-cophenolic acid (MPA) preparation, the average CYAtrough concentration target using the Abbot TDX assay(or equivalent) is 325 ng/ml (range, 275–375 ng/ml) forthe first 6 post-operative weeks, 275 ng/ml (range 200–350 ng/ml) for Weeks 6 to 12, 225 ng/ml (range 150–300 ng/mL) for Month 3 to Month 6, and 200 ng/ml(range 150–250 ng/mL) from Month 6 onwards.

Level of Evidence: C.. At present, CYA trough concentration targets when

CYA is used in combination with proliferation signalinhibitor (PSI; mammalian target of rapamycin [mTOR]inhibitors) agents have not been adequately determined.

Level of Evidence: C.. Measurement of 12-hour trough concentration for twice-

daily TAC and a 24-hour trough concentration for once-daily TAC is the recommended drug monitoring methodfor routine clinical use. The therapeutic range of TAClevels varies depending on concomitant drugs, toxicityconcerns, and time after HT. In general, when used inconjunction with AZA or a MPA preparation, TACtrough concentration targets range between 10 and 15ng/ml during the early post-operative period (Days0–60), between 8 and 12 ng/ml for the next 3 to 6months, and between 5 and 10 ng/ml in stable patients 6months after HT.

Level of Evidence: C.. At this time, target therapeutic TAC trough concentra-

tions when TAC is used in combination with PSI (mTORinhibitors) agents have not been adequately determined.

Level of Evidence: C.. Therapeutic drug monitoring for PSIs using trough con-

centration levels is recommended for sirolimus (SRL)and everolimus (EVL). Levels should be measured atleast 5 days after adjustment of the dose, when a newsteady state is achieved. When used in combination withCYA, the optimal trough target level for EVL is between3 and 8 ng/ml. The corresponding optimal trough levelfor SRL is 4 to 12 ng/ml.

Level of Evidence: B.

7

8

C

1

2

3

4

5

6

C

1

2

TR

RR(C

1

928 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

. In pediatric HT recipients, TAC and CYA should bemonitored using C0 levels when twice-daily dosing isused. Target levels are comparable to those in adults, butslightly lower targets may be used in low-risk patientssuch as non-sensitized infant HT recipients.

Level of Evidence: C.. There are insufficient data to support routine monitoring

of MPA levels in pediatric recipients. However, inter-mittent monitoring is reasonable when there is ongoingrejection, doubts about adequacy of dosing (eg, infantsand young children), and to assess medical compliance.

Level of Evidence: C.

lass IIb:

. At this time replacement of twice-daily TAC with once-daily TAC dosing cannot be recommended in HT recip-ients. Should a patient require the once-daily formula-tion, appropriate monitoring should be used to ensuremaintenance of appropriate levels and preserved heartallograft function.

Level of Evidence: C.. In patients with a therapeutic 12-hour trough concentra-

tion for twice daily TAC but evidence of potential drug-related toxicity or reduced efficacy (rejection), a 3-hourpost-dose level (C3) may help to adjust TAC doses.

Level of Evidence: C.. In selected situations (rejection, infection, renal failure,

malnutrition, and certain ethnic populations) where it issuspected that altered MMF exposure contributes toheart allograft dysfunction, measurement of trough MPAlevels may be used to guide drug dosing. In such cases,a MPA level of � 1.5 mg/liter is considered to besub-therapeutic.

Table 8 Significant Differences in Primary End Points betwee

Author (year) Study N Follow-up

Kobashigawa163 (1998) MMF vs AZA 650 3 years

Reichart209 (1998) TAC vs CYA 82 1 yearTaylor153 (1999) TAC vs CYA 85 1 yearEisen157 (2003) EVL vs AZA 634 1 year

Keogh156 (2004) SRL vs AZA 136 2 years

Grimm154 (2006) TAC vs CYA 314 1.5 year

Kobashigawa158 (2006) TAC/MMF vsTAC/SRL vsCYA/MMF

343 1 year

Baran159 (2007) TAC/MMF vs TAC 58 1 yearLehmkuhl160 (2008) EVL/rd-CYA vs

MMFsd-CYA176 1 year

CAV, cardiac allograft vasculopathy; CYA, cyclosporine; EVL, everolimmycophenolate mofetil; MMFsd, mycophenolate mofetil/standard exposu

aTreated-patient population (see text).bReanalysis of MMF IVUS data.209

Level of Evidence: C.

. Dose adjustments and frequency of therapy with poly-clonal antibodies (eg, anti-thymocyte globulin) used asinduction therapy can be monitored with daily measure-ment of CD3 or CD2 counts with the goal of maintainingthe CD2 or CD3 count between 25 and 50 cells/mm3 orabsolute total lymphocyte counts � 100 to 200 cells/mm3.

Level of Evidence: C.. In pediatric HT recipients CYA C2 monitoring may be

performed instead of C0 in centers with extensive expe-rience with this form of monitoring.

Level of Evidence: C.. As in adults, routine monitoring of SRL and EVL at C0

is recommended also in children.Level of Evidence: C.

lass III:

. Routine therapeutic drug monitoring of MPA levels toadjust MMF doses cannot be recommended at this time.

Level of Evidence: C.. Measuring CD 25 saturation to adjust the dose of anti-

interleukin-2 receptor antibodies remains experimentaland its routine clinical use cannot be recommended.

Level of Evidence: C.

opic 3: Principles of Immunosuppression andecommended Regimens

ecommendations on the Principles of Immunosuppressiveegimens in Heart Transplant Recipients150–210:See Table 8, Table 9A, Table 9B, and Table 10)lass I:

. Maintenance therapy should include a CNI in all pedi-

y Groups from Major Clinical Trials

al Rejection CAV by IVUS

higherivala

MMF � less rejection NS; MMF � less CAVat 1 yearb

NSNSEVL groups � less rejection EVL groups � less

CAVSRL groups � less rejection

at 6 monthsSRL groups � less

CAVTAC � less rejection at 6

months. . .

NS; TAC groups � lowerany-treated rejection

. . .

NS NSNS . . .

/rd, everolimus/reduced exposure; IVUS, intravascular ultrasound; MMF,not statistically significant; SRL, sirolimus; TAC, tacrolimus.

n Stud

Surviv

MMF �surv

NSNSNS

NS

NS

NS

NSNS

us; EVLre; NS,

atric HT recipients.

2

3

C

1

2

3

4

5

6

7

929Costanzo et al. Guidelines for Heart Transplant Care

Level of Evidence: C.. In adults, the use of statins beginning 1 to 2 weeks after

HT is recommended regardless of cholesterol levels.Owing to pharmacologic interactions with CNI and riskfor toxicity, initial statin doses should be lower thanthose recommended for hyperlipidemia.

Level of Evidence: A.. Creatinine kinase levels should be monitored in all chil-

dren receiving statins.Level of Evidence: C.

lass IIa:

. Calcineurin inhibitor-based therapy remains the standardin immunosuppressive protocols used after HT.

Level of Evidence: B.. MMF, EVL, or SRL as tolerated, should be included in

contemporary immunosuppressive regimens becausetherapies including these drugs have been shown to re-duce onset and progression of cardiac allograft vascu-lopathy (CAV) as assessed by intravascular ultrasound(IVUS).

Table 9 (A) Significant Differences in Adverse Events From th

First author (year) Study No. Renal fu

Kobashigawa163 (1998) MMF vs AZA 650

Reichart209 (1998) TAC vs CYA 82 NSTaylor153 (1999) TAC vs CYA 85 NS

Eisen157 (2003) EVL vs AZA 634 EVL grouworsefuncti

Keogh156 (2004) SRL vs AZA 136 SRL grouworsefuncti

Grimm154 (2006) TAC vs CYA 314 NS

Kobashigawa158 (2006) TAC/MMF vs TAC/SRLvs CYA/MMF

343 TAC/MMbest rfuncti

Baran159 (2007) TAC/MMF vs TAC 58 NS

Lehmkuhl160 (2008) EVL/rd-CYA vsMMFsd-CYA

176 NS

See Table 8 for abbreviations.

Level of Evidence: B.

. Immunosuppressive induction with polyclonal antibodypreparations may be beneficial in patients at high risk ofrenal dysfunction when used with the intent to delay oravoid the use of a CNI.

Level of Evidence: B.. In pediatric HT recipients, routine use of induction ther-

apy with a polyclonal preparation is indicated whencomplete CS avoidance is planned after HT.

Level of Evidence: C.. Routine use of statins is recommended for all pediatric

patients with evidence of hyperlipidemia, CAV, or afterretransplantation.

Level of Evidence: C.. TAC is the preferred CNI for pediatric HT recipients

considered at high immunologic risk (eg, sensitizedrecipients with evidence of donor-specific antibody[DSA]).

Level of Evidence: C.. CS avoidance, early CS weaning, or very low dose main-

tenance CS therapy are all acceptable therapeutic ap-proaches.

r Clinical Trials

InfectionsCholesterol &triglycerides Hypertension

MMF � moreanyopportunisticinfection

NS CYA � more hypertensionNS CYA �

higherchol & tri

CYA � more hypertension

EVL groups �lower viral/CMV but morebacterialinfections

EVL groups� higherchol & tri

NS

SRL groups �lower CMV butmorepneumonia

NS for chol;SRLgroups �highertrig

NS

NS CYA �higherchol & tri

CYA � more hypertension

TAC/SRL �lower viral butmore fungalinfections

NS for chol;TAC/MMF� lowertrig

NS

TAC/MMF �morehospitalizedinfections

. . . . . .

EVL � Less CMVinfections

. . . . . .

e Majo

nction

ps �renalon

ps �renalon

F �enalon

Level of Evidence: B.

8

9

C

1

2

3

4

5

6

7

8

930 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

. If used, CS weaning should be attempted if there aresignificant CS side effects and no recent rejection epi-sodes (eg, within 6 months).

Level of Evidence: C.. Pediatric recipients with pre-formed alloantibodies and a

positive donor-specific cross-match should receive in-duction therapy, and TAC-based “triple therapy” withCSs and either MMF or an mTOR inhibitor.

Level of Evidence: C.

lass IIb:

. The results of clinical trials suggest that TAC-basedregimens may be associated with lower rejection ratesbut not with superior survival after HT than CYA-based regimens.

Level of Evidence: B.

Table 9 (B) Significant Differences in Adverse Events From th

First author (year) Study N Hematologi

Kobashigawa163 (1998) MMF vs AZA 650 AZA � mor

Reichart209 (1998) TAC vs CYA 82 . . .

Taylor153 (1999) TAC vs CYA 85 NSEisen157 (2003) EVL vs AZA 634 NSKeogh156 (2004) SRL vs AZA 136 SRL groups

& thromb

Grimm154 (2006) TAC vs CYA 314 TAC � more

Kobashigawa158 (2006) TAC/MMF vs TAC/SRLvs CYA/MMF

343 NS

Baran159 (2007) TAC/MMF vs TAC 58 NSLehmkuhl160 (2008) EVL/rd-CYA vs

MMFsd-CYA176 MMF � mor

See Table 8 for abbreviations.

Table 10 Recommendation for Statin Doses in HeartTransplant Patients201,202,210,387–393

Drug Dose Risks

Pravastatin 20–40 mg394 Myositis (lower)Simvastatin 5–20 mg395 Myositis (higher)

� 20 mg not recommendedAtorvastatin 10–20 mg387 Myositis (higher)Fluvastatin 40–80 mg389 Myositis (lower)Lovastatin 20 mg390 Myositis (higher)Rosuvastatin 5–20 mg388 Myositis

. The adverse events of immunosuppressive drugs observedin randomized clinical trials underscore the need for indi-vidualization of immunosuppression according to the char-acteristics and risks of the individual HT recipient.

Level of Evidence: C.. Most children should receive adjunctive therapy with an

anti-metabolite or a PSI.Level of Evidence: C.

. If a child is intolerant of adjunctive therapy, the decisionwhether or not to replace it with another agent should bemade after review of the patient’s rejection history andimmunologic risk. TAC monotherapy is acceptable inpatients with a benign rejection history.

Level of Evidence: C.. For children diagnosed with CAV, the addition of an

mTOR inhibitor should be strongly considered.Level of Evidence: C.

. Routine use of immunosuppressive induction in all pa-tients has not been shown to be superior to immunosup-pressive regimens that do not use such therapy.

Level of Evidence: B.. Immunosuppressive induction with anti-thymocyte glob-

ulin (ATG) may be beneficial in patients at high risk foracute rejection.

Level of Evidence: C.. Routine use of statins is recommended for adolescents

and selected younger children with at an increased risk ofrejection or CAV.

r Clinical Trials

GI Disorders Other

penia MMF � more diarrhea andesophagitis

NS for hyperglycemiatreatment

. . . NS for glucoseintolerance

. . . . . .NS NS for wound infection

re anemiaenia

AZA � more nausea; SRLgroups � more diarrhea

AZA � more arrhythmiaand atrial fibrillation;SRL groups � moremouth ulcers &abnormal healing

ia CYA � more cholelithiasis TAC � more diabetesmellitus & tremor;CYA � more gumhyperplasia &hirsutism

. . . TAC/SRL � moreinsulin therapy &impaired woundhealing; NS fordiabetes mellitus

. . . NS for malignancyopenia . . . . . .

e Majo

c

e leuko

� moocytop

anem

e leuk

Level of Evidence: C.

T

RC(C

1

2

3

4

5

6

7

8

9

1

1

RAC

1

2

3

4

5

C

1

2

931Costanzo et al. Guidelines for Heart Transplant Care

opic 4: Treatment of Acute Cellular Rejection

ecommendations for Treatment of Symptomatic Acuteellular Rejection205–207,211–217:See Table 11)lass I:

. An EMB should be performed as early as possible ifthere is suspicion of symptomatic acute heart allograftrejection.

Level of Evidence: C.. The HT recipient with symptomatic acute cellular rejec-

tion should be hospitalized. Patients with hemodynamiccompromise should be treated in the ICU.

Level of Evidence: C.. High-dose IV CS should be first-line therapy for symp-

tomatic acute cellular rejection irrespective of ISHLTEMB grade (1R, 2R or 3R).

Level of Evidence: C.. Cytolytic immunosuppressive therapy with anti-thymo-

cyte antibodies should be administered in addition to IVCS if hemodynamic compromise is present, and espe-cially if there is no clinical improvement within 12 to 24hours of IV CS administration.

Level of Evidence: C.. IV inotropes and vasopressors should be used as nec-

essary to maintain adequate CO and systemic bloodpressure until recovery of heart allograft function oc-curs.

Level of Evidence: C.. Anti-microbial prophylaxis against opportunistic in-

fections should be administered when high-dose CSand/or cytolytic therapy are used for the treatment ofrejection.

Level of Evidence: C.. Appropriate adjustments of maintenance immunosup-

pressive therapy should be made to decrease the risk

Table 11 Suggested Dosing of Medications Used forTreatment of Acute Cellular Rejection

Medication Dose Duration

CorticosteroidsMethylprednisolone

(high-dose)250–1000 mg/day IV 3 daysa

Prednisone 1–3 mg/kg/day PO 3–5 daysa

Polyclonal anti-thymocyteantibody

Thymoglobulinb 0.75–1.5 mg/kg/day 5–14 daysATGAMb 10 mg/kg/day 5–14 daysATG-Freseniusb 3 mg/kg/day 5–14 days

Monoclonal antibody 5 mg/day 5–14 daysMuromonoab-

CD3 (OKT3)b

ATG, anti-thymocyte gamma-globulin-fresenius; ATGAM, anti-thy-mocyte gamma-globulin; IV, intravenous; PO, oral (per os).

aCorticosteroid taper can be considered.bPremedicate with CS, anti-histamine and anti-pyretic.

of recurrent rejection. These can include ascertain-

ment of compliance with current therapy, increase inthe dose of current immunosuppressive agent(s), ad-dition of new agent(s), or conversion to differentagent(s).

Level of Evidence: C.. Follow-up EMB should be done 1 to 2 weeks after

initiation of therapy for acute cellular rejection.Level of Evidence: C.

. Serial echocardiograms should be used to monitorchanges in heart allograft function in response to anti-rejection therapy.

Level of Evidence: C.0. In a patient with low-grade acute cellular rejection and

hemodynamic compromise, the possibility of AMRshould also be entertained (see AMR section).

Level of Evidence: C.1. Interleukin-2 receptor blockers should not be used to

reverse acute cellular rejection.Level of Evidence: C.

ecommendations for the Treatment of Asymptomaticcute Cellular Rejection:205–207,211–217

lass I:

. Severe acute cellular rejection (ISHLT 3R) diagnosedby surveillance EMB should be treated even in theabsence of symptoms or evidence of heart allograftdysfunction.

Level of Evidence: C.. High dose IV CS should be given for asymptomatic

severe (ISHLT 3R) acute cellular rejection.Level of Evidence: C.

. Asymptomatic moderate acute cellular rejection (ISHLT2R) can be treated with either IV or oral CS.

Level of Evidence: C.. Adjustment of maintenance immunosuppressive therapy

should be done in patients with asymptomatic moderate(ISHLT 2R) or severe (ISHLT 3R) acute cellular rejec-tion. This can include an increase of the dose of currentmedications, addition of an agent, or conversion to adifferent maintenance regimen.

Level of Evidence: C.. Anti-microbial prophylaxis against opportunistic infec-

tions should be administered when high-dose CSs and/orcytolytic therapy are used for treatment of rejection..

Level of Evidence: C.

lass IIa:

. The performance of a follow-up EMB should be consid-ered 2 to 4 weeks after initiation of therapy for asymp-tomatic moderate or severe acute cellular rejection.

Level of Evidence: C.. Cytolytic immunosuppressive therapy can be considered

if there is no histologic resolution of rejection on thefollow-up EMB.

Level of Evidence: C.

3

C

1

RAC

1

2

3

C

1

2

TM

RtC

1

1

C

1

Rj(C

1

2

932 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

. Asymptomatic mild cellular rejection (ISHLT 1R) doesnot require treatment in the vast majority of cases.

Level of Evidence: C.

lass IIb:

. Asymptomatic moderate cellular rejection (ISHLT2R), especially if occurring later than 12 months afterHT, may not require treatment. Close surveillance(clinical, echocardiographic, and follow-up EMB) isstrongly suggested if no treatment is administered inthis setting.

Level of Evidence: C.

ecommendations for Treatment of Recurrent or Resistantcute Cellular Rejection:218,219

lass I:

. For recurrent or CS-resistant acute cellular rejection,cytolytic immunosuppressive therapy with anti-thymo-cyte antibodies should be considered.

Level of Evidence: C.. Maintenance immunosuppression should be re-evaluated

in patients with recurrent/resistant HT rejection (seeabove).

Level of Evidence: C.. Frequent surveillance of heart allograft function (eg, by

echocardiography) is recommended in patients with re-current/resistant rejection, even if persistently asymp-tomatic.

Level of Evidence: C.

lass IIb:

. Additional approaches that can be considered for recur-rent or resistant acute cellular rejection include metho-trexate pulse therapy, photopheresis, and total lymphoidirradiation.

Level of Evidence: B.. Evaluation of EMB specimens for concomitant anti-

body-mediated rejection (AMR; see the Recommen-dations for Treatment of Antibody-Mediated Rejec-tion) and determination of the presence of anti-HLAantibodies in the HT recipient’s serum is also sug-gested.

Table 12 Examples of Therapies for Antibody-Mediated Rejec

Therapeutic modality Dose

Plasmapheresis 1–2 plasma exchanges

IV Ig 100–1,000 mg/kg

Rituximab 375 mg/m2

IV Ig, intravenous immunoglobulin.Based on Grauhan O et al,221 Leech SH et al,226 Michaels PJ et al,228 M

Level of Evidence: C.

opic 5: Treatment of Hyperacute and Antibody-ediated Rejection

ecommendations for the Treatment of Hyperacute Rejec-ion:25,220

lass I:

. Treatment for hyperacute rejection should be initiated assoon as the diagnosis is made, preferably when the HTrecipient is still in the operating room. Treatments thatshould be considered include (1) high-dose IV CS; (2)plasmapheresis; (3) IV immunoglobulin; (4) cytolyticimmunosuppressive therapy; (5) IV CNI (CYA, TAC)and metabolic cycle inhibitors (MMF); (6) IV inotropesand vasopressors; and (7) MCS.

Level of Evidence: C.

. Intraoperative myocardial EMB should be obtained toconfirm the diagnosis of hyperacute heart allograft rejec-tion.

Level of Evidence: C.

lass IIb:

. Urgent retransplantation may be considered if the abovemeasures do not result in restoration of acceptable heartallograft function, but repeat HT in the setting of hyper-acute rejection is associated with high mortality.

Level of Evidence: C.

ecommendations for Treatment of Antibody Mediated Re-ection85,221–229:See Table 12)lass IIa:

. The following treatments can be used to disrupt theimmune-mediated injury of the heart allograft in AMR:(1) high-dose IV CS and (2) cytolytic immunosuppres-sive therapy.

Level of Evidence: C.. The following treatments may be used to remove circu-

lating anti-HLA antibodies or decrease their reactivity:(1) plasmapheresis; (2) immune apheresis (immunoad-

Frequency Duration

Daily 3–5 daysEvery other day 1–2 weeks3 times per week 1–4 weeksOnce weekly 2–4 weeks1–3 times per week, often given

after each plasmapheresis1–4 weeks

Once weekly 1–4 weeks

et al,229 Kaczmarek I et al,222 Takemoto SK et al,85 and Bierl C et al.396

tion

iller LW

sorption); and (3) IV Ig.

3

4

5

6

7

C

1

2

T

RtC

1

C

1

2

3

4

5

C

1

TT

C

MRGGJ

T

RpC

1

2

C

1

933Costanzo et al. Guidelines for Heart Transplant Care

Level of Evidence: C.. The following treatments are used to maintain adequate

cardiac output and systemic blood pressure: (1) IV ino-tropes and vasopressors and (2) MCS.

Level of Evidence: C.. When AMR is suspected, EMB examination should be

expanded to include immunohistochemistry stains forcomplement split products and possibly antibody.

Level of Evidence: C.. Recipient serum should be screened for presence, quan-

tity, and specificity of anti-donor (HLA) antibodies.Level of Evidence: C.

. Follow-up EMB should be performed 1 to 4 weeks afterinitiation of therapy and include immunohistochemistryexamination.

Level of Evidence: C.. Adjustment of maintenance immunosuppressive therapy

may be considered. This can include increase in the doseof current immunosuppressive agent(s), addition of newagent(s), or conversion to different agent(s).

Level of Evidence: C.

lass IIb:

. Systemic anti-coagulation may decrease intravascularthrombosis in the heart allograft.

Level of Evidence: C.. Emergent retransplantation may be considered if the

above measures do not restore acceptable heart allograftfunction, but outcomes in this situation are unfavorable.

Level of Evidence: C.

opic 6: Management of Late Acute Rejection

ecommendation for the Management of Late Acute Rejec-ion:230,231

lass I:

. Maintenance immunosuppression and the intensity ofclinical follow-up should be reevaluated after symptom-atic or asymptomatic late acute heart allograft rejection.

Level of Evidence: C.

lass IIa:

. After the first year, EMB surveillance (eg, every 4–6months) for an extended period of time is recommendedin patients at higher risk for late acute rejection, toreduce the risk of rejection with hemodynamic compro-mise, and to reduce the risk of death in African-Ameri-can recipients.

Level of Evidence: C.. Repeated education on the critical importance of adher-

ence to treatment, and early reporting of symptoms con-tribute to the prevention and early recognition of lateacute rejection.

Level of Evidence: C.. Patients at low risk for late rejection do not appear to

significantly benefit from indefinite EMB surveillance.The usefulness of long-term routine EMB should be

evaluated against the risks and the costs of the procedure.

Repeated EMB increase the probability of damage to theTV apparatus and collection of non-diagnostic material.

Level of Evidence: C.. In pediatric HT recipients, CAV should be considered in

the differential diagnosis of late symptomatic or asymp-tomatic rejection when heart allograft dysfunction ispresent. Coronary angiography (and possibly IVUS)should be considered in these patients.

Level of Evidence: C.. In pediatric HT recipients, late rejection has negative

prognostic implications and may be associated with anincreased risk for subsequent development of CAV; con-sequently, a follow-up coronary angiography may berecommended.

Level of Evidence: C.

lass IIb:

. In pediatric HT recipients, withholding treatment forasymptomatic mild-moderate late heart allograft rejec-tion is reasonable but requires close follow-up.

Level of Evidence: C.

ask Force 3: Long-term Care of Heartransplant Recipients

hair: Sharon Hunt, MD; Co-Chair: Michael BurchContributing Writers: Geetha Bhat, MD; Charles Canter,

D; Richard Chinnock, MD; Marisa Crespo-Leiro, MD;eynolds Delgado, MD; Fabienne Dobbels, PhD; Kathleenrady, PhD; Walter Kao, MD; Jaqueline Lamour, MD;areth Parry, MD; Jignesh Patel, MD; Daniela Pini, MD;

effrey Towbin, MD; Gene Wolfel, MD

opic 1: Minimization of Immunosuppression

ecommendations for the Minimization of Immunosup-ression:159,162,188,232–242

lass I:

. CS withdrawal can be successfully achieved 3 to 6months after HT in many low-risk patients (those with-out circulating anti-HLA antibodies, non-multiparouswomen, those without a history of rejection, and olderHT recipients).

Level of Evidence: B.. Lower levels of CNI in HT recipients should be sought

when CNI are used in conjunction with MMF (comparedwith AZA) because with this combination lower levelsare safe and associated with lower rejection rates as wellas improved renal function.

Level of Evidence: B.

lass IIa:

. A PSI may be substituted for CNI later than 6 monthsafter HT to reduce CNI-related nephrotoxicity and CAVin low-risk recipients.

Level of Evidence: C.

C

1

2

3

4

C

1

2

TA

RpC

1

2

3

C

1

T

RC(C

1

2

3

4

5

934 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

lass IIb:

. CNI monotherapy with early CS withdrawal may beconsidered in highly selected individuals. This strategyhas been associated with acceptable short-term outcomesin HT recipients.

Level of Evidence: B.. In pediatric HT recipients, minimization of immunosup-

pression by CS withdrawal is common practice and ap-pears safe, with the majority of children being free of CSby 5 years after HT.

Level of Evidence: C.. Due to variable pharmacokinetics in children, strategies

for minimization of immunosuppression in the pediatricpopulation may require a greater reliance on drug levelsmonitoring than in adults.

Level of Evidence: C.. The use of PSI may be considered in pediatric HT re-

cipients to reduce CAV and nephrotoxicity, but insuffi-cient data are available on the effects of PSI in children.

Level of Evidence: C.

lass III:

. In HT recipients, substitution of PSI for MMF for the specificpurpose of lowering CNI exposure to reduce CNI-relatednephrotoxicity is not recommended due to the interaction be-tween CNI and PSI, which enhances CNI nephrotoxicity.

Level of Evidence: C.. Substitution of a PSI for MMF earlier than 3 months

after HT is not recommended due to a higher risk ofrejection as well as delayed wound healing.

Level of Evidence: B.

opic 2: Management of Neurologic Complicationsfter Heart Transplantation

ecommendations for the Management of Neurologic Com-lications After Heart Transplantation:243–246

lass I:

. Management of HT recipients with seizures should in-clude reduction of CNI doses (taking into considerationthe risk of inadequate immunosuppression) and correc-tion of hypomagnesemia, if present.

Level of Evidence: C.. The occurrence of encephalopathy late after HT should

prompt neurologic consultation and imaging to identifypossible underlying etiologies.

Table 13 Basic Criteria for the Interpretation of Intravascula

Normal

Baseline study (4–6 weeks after hearttransplantation)

0.25–0.50 mm intimal

Study 1 year after heart transplantation No change in intimal t

Level of Evidence: C.

. Posterior reversible leukoencephalopathy (PRES) inHT recipients should be managed with a reduction ofCNI doses or substitution with an alternative CNI.

Level of Evidence: C.

lass IIb:

. Heart transplant recipients who continue to experienceseizures after a reduction in the CNI dose may benefitfrom CNI withdrawal and substitution with a PSI (SRL,EVL).

Level of Evidence: C.

opic 3: Cardiac Allograft Vasculopathy

ecommendations for the Diagnosis and Management ofardiac Allograft Vasculopathy155–157,164,247–267

See Table 13)lass I:

. Primary prevention of CAV in HT recipients shouldinclude strict control of cardiovascular risk factors (hy-pertension, diabetes, hyperlipidemia, smoking, and obe-sity) as well as strategies for the prevention of CMVinfection.

Level of Evidence: C.. In HT recipients, statin therapy has been shown to reduce

CAV and improve long-term outcomes regardless oflipid levels and should be considered for all HT recipi-ents (adult and pediatric).

Level of Evidence: A.. Annual or biannual coronary angiography should be con-

sidered to assess the development of CAV. Patients freeof CAV at 3 to 5 years after HT, especially those withrenal insufficiency, may undergo less frequent invasiveevaluation.

Level of Evidence: C.. Follow-up coronary angiography is recommended at

6 months after a percutaneous coronary interven-tion because of high restenosis rates in HT reci-pients.

Level of Evidence: C.. Selective coronary angiography is the investigation of

choice for the diagnosis of CAV in pediatric HT recip-ients. It should be performed at yearly or biannual inter-vals.

sound Measurements After Heart Transplantation

Abnormal

ess Any intimal lesion � 0.5 mm suggests donor disease247

ss An increase in intimal thickness � 0.5 mm from baselinesuggests accelerated CAV associated with adverseoutcomes249

r Ultra

thickn

hickne

Level of Evidence: C.

C

1

2

3

4

5

6

7

8

9

1

C

1

2

T

RHC

1

2

3

4

C

1

TT

RTC

1

2

3

935Costanzo et al. Guidelines for Heart Transplant Care

lass IIa:

. A baseline coronary angiogram at 4 to 6 weeks after HTmay be considered to exclude donor coronary arterydisease.

Level of Evidence: C.. IVUS in conjunction with coronary angiography with a

baseline study at 4 to 6 weeks and at 1 year after HT isan option to exclude donor coronary artery disease, todetect rapidly progressive CAV, and provide prognosticinformation.

Level of Evidence: B.. In HT recipients with established CAV, the substitution

of MMF or AZA with a PSI can be considered.Level of Evidence: B.

. A PSI can been used in pediatric HT recipients whodevelop CAV, but the effect of PSI on the progression ofCAV in children is unknown.

Level of Evidence: C.. IVUS can be safely used in older pediatric HT recipients

to assess CAV.Level of Evidence: C.

. Evaluation of coronary flow reserve in conjunction withcoronary angiography may be useful for the detection ofsmall-vessel coronary disease, which is a manifestationof CAV.

Level of Evidence: C.. Treadmill or dobutamine stress echocardiography and

myocardial perfusion imaging may all be useful for thedetection of CAV in HT recipients unable to undergoinvasive evaluation. Non-invasive testing for CAV istechnically possible in children.

Level of Evidence: B.. Percutaneous coronary intervention with drug-eluting

stents is recommended in both adults and children withCAV and offers short-term palliation for appropriatediscrete lesions.

Level of Evidence: C.. Surgical revascularization in HT recipients with CAV is

an option in highly selected patients who have lesionsamenable to surgical revascularization.

Level of Evidence: C.0. Cardiac retransplantation may be considered in patients

with severe CAV and absence of contraindications forrepeat HT.

Level of Evidence: C.

lass IIb:

. Ultrafast computed tomography (CT) for the detection ofcoronary calcium has been used mostly as an investiga-tional tool for assessing CAV in HT recipients, but isbeing superseded by advances in CT angiography.

Level of Evidence: C.. CT coronary angiography shows promise in the evalua-

tion of CAV in HT recipients, although higher restingheart rates in these patients limit the technical quality ofthis study.

Level of Evidence: C.

opic 4: Malignancy After Heart Transplantation

ecommendations on the Approach to Malignancy Aftereart Transplantation:268–271

lass I:

. Recommendations regarding screening for breast, colon,and prostate cancer in the general population should alsobe followed in HT recipients.

Level of Evidence: C.. It is recommended that HT recipients have close skin

cancer surveillance, including education on preventivemeasures and yearly dermatologic examinations.

Level of Evidence: C.. Initial evaluation and a therapeutic plan for post-trans-

plant lymphoproliferative disorder (PTLD) in HT recip-ients should be done at the transplant center by physi-cians familiar with transplant-associated malignancies.

Level of Evidence: C.. There is no evidence to support a reduction in immuno-

suppression in patients with solid tumors unrelated to thelymphoid system. Maintenance immunosuppressionshould be continued unless there are specific reasons toreduce certain drugs, such as reduction of bone marrow-suppressive agents if leucopenia occurs.

Level of Evidence: C.

lass IIa:

. Chronic immunosuppression should be minimized in HTrecipients as possible, particularly in patients at high riskfor malignancy.

Level of Evidence: C.

opic 5: Chronic Kidney Disease After Heartransplantation

ecommendations on Chronic Kidney Disease After Heartransplantation:70,242–244,272–282

lass I:

. Estimation of glomerular filtration rate (GFR) with themodified diet in renal disease (MDRD) equation, urinal-ysis, and spot urine albumin/creatinine ratio should beobtained at least yearly after HT. Measurement of sCr forestimation of GFR should be obtained more often inpatients with GFR � 60 ml/min/1.73 m2, and/or fastGFR decline in the past (� 4 ml/min/1.73 m2 per year).

Level of Evidence: C.. Although in children there is no consensus on the opti-

mal method to estimate GFR, this measurement shouldbe done and a urinalysis obtained at least yearly inpediatric HT recipients.

Level of Evidence: C.. Heart transplant recipients with an estimated GFR � 30

ml/min/1.73 m2, proteinuria � 500 mg/day (or urinealbumin/creatinine ratio � 500 mg/g), or rapidly declin-ing GFR (� 4 ml/min/1.73 m2 per year), should be

referred to a nephrologist for management of metabolic

4

5

6

7

8

9

1

C

1

TH

RHC

1

2

3

4

5

6

7

8

C

1

TI

Rc(C

1

936 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

abnormalities and other complications of renal insuffi-ciency and consideration of renal transplantation.

Level of Evidence: C.. In all HT recipients (adult and pediatric) with chronic

kidney disease (CKD), CNI exposure should be loweredto the minimum level required for effective immunosup-pression. In patients taking AZA, this may be achievedby conversion of AZA to MMF.

Level of Evidence: B.. Owing to the potential for precipitating rejection, CNI-

free regimens should be used with caution in HT recip-ients with significant renal insufficiency that persistsdespite CNI reduction.

Level of Evidence: C.. In pediatric HT recipients, CS minimization or with-

drawal should be attempted to avoid hypertension andsubsequent CKD, as long as there is no clinical rejection.There are no strong data in adult HT recipients.

Level of Evidence: B.. Interventions that have been proven to slow the progres-

sion of CKD in the general population should be con-sidered in all HT recipients. These include strict glucoseand blood pressure control and use of an angiotensin-converting enzyme inhibitor (ACEI) or angiotensin re-ceptor blocker (ARB). The American Diabetes Associa-tion (ADA) or the International Diabetes FederationGuidelines should be used to manage diabetes. Bloodpressure should be treated according to the Joint NationalCommittee VII or the European Society of Cardiology2007 Guidelines.

Level of Evidence: C.. In pediatric HT recipients, diabetes is rare. In contrast,

hypertension is common, and adequate blood pressurecontrol with a CCB or ACEI is warranted to avoid CKD.

Level of Evidence: C.. Hemoglobin (Hgb) levels should be measured at least

annually in all HT patients with CKD. If anemia (Hgb �13.5 g/dl in adult men; � 12 g/dl in adult women) isdetected, iron status should be addressed and erythropoi-esis-stimulating agents should be used to maintain Hgblevels between 11 and 13 g/dl.

Level of Evidence: C.0. Kidney transplantation should be considered the treat-

ment of choice for all HT recipients (adult and pediat-ric) with end-stage renal disease who are appropriatecandidates. Living donation should be considered.

Level of Evidence: C.

lass IIa:

. CCBs should be considered the anti-hypertensive drug ofchoice when optimal blood pressure control cannot beachieved with ACEI/ARB or when these drugs are con-traindicated in HT recipients.

Level of Evidence: C.

opic 6: Management of Diabetes Mellitus After

eart Transplantation

ecommendations for the Management of Diabetes AfterT:283–288

lass I:

. Prevention, early detection, and appropriate therapy fordiabetes should be considered as an important compo-nent of patient care after HT.

Level of Evidence: C.. Patients should be periodically screened for diabetes

after HT by measuring fasting plasma glucose levelsor with an oral glucose tolerance test (more sensitivescreening test for pre-diabetic state) and HgbA1C de-termination, as appropriate. The frequency of screen-ing will depend on risk factors and immunosuppres-sive therapy.

Level of Evidence: C.. Therapies for short-term peri-operative and long-term

chronic glycemic control in HT recipients should bebased on ADA recommendations.

Level of Evidence: C.. Heart transplant recipients with diabetes should be coun-

seled regarding weight control, diet and nutrition, andexercise.

Level of Evidence: C.. Pre-HT risk factors should be assessed, and diabetogenic

immunosuppressive medications should be minimizedwhenever possible in HT recipients.

Level of Evidence: C.. CS-sparing regimens and decreased CNI doses should be

used as appropriate to prevent diabetes in HT recipients.Level of Evidence: C.

. Associated cardiovascular risk factors (in addition todiabetes), such as hyperlipidemia and hypertension,should be managed aggressively in HT recipients. An-nual measurements of lipids levels should be performedaccording to ADA recommendations.

Level of Evidence: C.. Annual screening should be performed for diabetic com-

plications (ophthalmology, podiatry, peripheral vasculardisease, etc) in HT recipients with diabetes.

Level of Evidence: C.

lass IIa:

. An endocrinology consultation may be considered whena pre-diabetic state or diabetes is diagnosed in a HTrecipient.

Level of Evidence: C.

opic 7: Other Complications of Chronicmmunosuppression

ecommendations on the Management of Various Compli-ations of Chronic Immunosuppression289–333:See Table 14)lass I:

. Recommendations for addressing other complicationsof immunosuppression include regular screening for

adverse events, minimizing drug doses, drug substitu-

T

RtC

1

937Costanzo et al. Guidelines for Heart Transplant Care

tion, and drug withdrawal (as previously discussed),as well as initiating targeted therapies for a specificcomplication. For example, anti-hyperuricemic ther-apy and concurrent risk reduction may be used toprevent recurrent attacks of gout, whereas acquiredcataracts require surgical intervention. It is importantto assess for contraindications and drug interactionswhen medically treating complications of immunosup-pression.

Table 14 Complications of Immunosuppressive Drugs

Drug Toxicities

Calcineurin inhibitors: cyclosporineand tacrolimus

Cardiovascular: hypertenNeurologic: headache, tr

syndrome ParkinsonismHematologic: Anemia, leDermatologic: fibrovascuGastrointestinal: nausea,

malakoplakia, eosinopveno-occlusive disease

Endocrine/metabolic: hyphyperlipemia311

Renal: renal dysfunctionInfection311

Mammalian target of rapamycininhibitors

Cardiovascular: edema, hNeurologic: Headache, pHematologic: Anemia, th

thromboses298,311,319,3

Respiratory: Dyspnea, pualveolar hemorrhage29

Endocrine and metabolicDermatologic: Acneiform

healing complicationsMusculoskeletal: extremi

impaired wound healinGastrointestinal: Diarrhea

hepatotoxicity311,325,3

Genitourinary: urinary traMycophenolate mofetil Infection (eg, herpes sim

Gastrointestinal (eg, naudistension and pain, e

Metabolism and nutritionCardiovascular (eg, hypeHematologic (eg, leukopNervous system (eg, heaRespiratory (eg, dyspneaRenal (eg, increased BUDermatologic (eg, rash)3

Corticosteroids (CS) Gastrointestinal (eg, pepNeuromuscular and skele

myopathy)311,330–333

Central nervous system (Dermatologic (eg, bruisinEndocrine and metabolic

growth suppression inunresponsiveness in ti

Ocular complications (eg

BOOP, bronchiolitis obliterans with organizing pneumonia; BUN, blo

Level of Evidence: C.

opic 8: Hypertension After Heart Transplantation

ecommendations on the Management of Hypertension Af-er Heart Transplantation:334–339

lass I:

. Because anti-hypertensive therapy in HT recipients hasbenefits similar to those in the general population, hy-pertension after HT should be treated to achieve the same

dema311

insomnia, hearing loss posterior reversible encephalopathytral and peripheral neuropathy, seizures303–311

ia, thrombotic microangiopathy, eosinophilia294,311,312

yps alopecia,295,313 hirsutism, gingival hyperplasia296

ea, steatohepatitis, cholestatic jaundice, colonicastroenterocolitis, villous atrophy/food allergies, hepatic,311,314–317

phatemia, hypomagnesemia, hyperglycemia, hyperkalemia,

opathy311

nsion311

ive multifocal encephalopathy, optic neuropathy311,318

cytopenia, thrombotic microangiopathy, venous

ry toxicity, interstitial pneumonitis, c, alveolar proteinosis,1,321,322

rtriglyceridemia, hypercholesterolemia311

dermatitis, ulcerating rash: perforating collagenosis, woundcence, leukocytoclastic vasculitis301,323

hedema (bilateral and unilateral); lingual angioedema;24

ea, vomiting, gastroduodenal ulcer disease;

ection, infertility (oligospermia)302,311,327

irus and cytomegalovirus)291,328,329

nstipation, diarrhea, vomiting, dyspepsia, abdominalgitis)291,328,329

, hyperglycemia, hypercholesterolemia, gout)291,311

n, peripheral edema)291,311

hrombocytopenia)291,311,328

tremor)291,311

ratory tract infection, cough)291

or creatinine)311

er, esophagitis, pancreatitis)311

, osteoporosis, pathologic fractures, muscle mass loss, CS

otional instability, headache)311

n fragile skin, impaired wound healing)311

ements (eg, diabetes mellitus, hyperlipidemia, fluid retention,en, adrenal suppression, adrenocortical and pituitaryf stress, and menstrual irregularities)311

oma, cataracts)311

nitrogen.

sion, eemor,, cen

ukopenlar poldiarrh

hilic g294,297

ophos

/nephr

yperterogressrombo20

lmona9,300,31

: Hypefacial

: dehisty lympg289,3

, naus26

ct infplex v

sea, cosophaal (egrtensioenia, tdache,, respiN and/11

tic ulctal (eg

eg, emg, thiderangchildrmes o, glauc

od urea

goals recommended for the general population.

2

3

4

5

C

1

TB

G

apid

sAospiotmia

nfsd

RIC

1

2

3

4

5

6

7

8

9

1

C

1

2

938 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

Level of Evidence: C.. Lifestyle modifications, including weight loss, low-so-

dium diet, and exercise are appropriate adjuncts to facil-itate control of blood pressure in HT recipients.

Level of Evidence: C.. Drug choice for treatment of hypertension in HT recip-

ients is empiric and depends on blood pressure re-sponses. CCBs are most widely used, but ACEI andARB may be preferred in diabetic recipients, and a2-drug regimen can include both CCB and ACEI/ARB.

Level of Evidence: C.. Modification of risk factors such as diabetes and hyper-

lipidemia are appropriate as adjunctive treatment forhypertension in HT recipients.

Level of Evidence: C.. Appropriate adjustment of immunosuppressive therapy,

especially CS weaning, may be helpful in managementof hypertension in HT recipients.

Level of Evidence: C.

lass IIa:

. Hypertension is common in both adults and childrenafter HT and can be assessed with ambulatory bloodpressure monitoring.

Level of Evidence: C.

opic 9: Prophylaxis for Corticosteroid-Inducedone Disease

aps in Evidence:Bisphosphonates continue to suppress bone reabsorption

fter discontinuation of therapy. It is not known, however, ifre-operative administration of these drugs can prevent thencreased bone loss that develops after HT with the intro-uction of CS.

Gaps in Evidence340:The predictive role of bone mass density (BMD) mea-

urement for fracture risk is unproven in HT recipients.lthough several studies have described a beneficial effectf bisphosphonates and vitamin D analogues on bone den-ity in adult HT recipients, none of these studies has beenowered to detect a decrease in fracture rate. In addition,mportant issues that remain unresolved include which is theptimal bisphosphonate, the route and duration of adminis-ration, and whether therapy should be continuous or inter-ittent. More research is also needed to define appropriate

ndications for bisphosphonate therapy and the optimalgent, dose, and duration of use in pediatric patients.

The potential role in the HT population of the recombi-ant human parathyroid hormone (teriparatide), a boneorming agent, and strontium ranelate, the first agent totimulate bone formation while decreasing reabsorption,eserves investigation.

ecommendations for the Prophylaxis of Corticosteroid-nduced Bone Disease After Heart Transplantation:341–351

lass I:

. All adult HT candidates should be screened for pre-existing bone disease, preferably at the time of place-ment on the waiting list. In adults, baseline BMD shouldbe obtained with a dual energy x-ray absorptiometry(DEXA) scan of the lumbar spine and femoral neck.

Level of Evidence: C.. The presence of low BMD or vertebral fractures should

prompt evaluation and treatment of correctable second-ary causes of osteoporosis, because significant improve-ment in BMD can be attained during the waiting periodfor HT. Bisphosphonates should be considered the treat-ment of choice.

Level of Evidence: C.. All HT candidates and recipients should have the rec-

ommended daily allowance for calcium (1,000–1,500mg, depending on age and menopausal status) and vita-min D (400–1,000 IU, or as necessary to maintain serum25-hydroxyvitamin D levels above 30 ng/ml � 75 nmol/L).

Level of Evidence: C.. After HT, regular weight-bearing and muscle-strength-

ening exercises should be encouraged to reduce the riskof falls and fractures and to increase bone density.

Level of Evidence: B.. In pediatric HT recipients, it is important to monitor

growth and pubertal development and be alert to thedevelopment of signs and symptoms of bone disease.

Level of Evidence: C.. Reduction or withdrawal of CS in pediatric HT recipi-

ents should be considered in the absence of precedingrejection with close monitoring for clinical rejection.

Level of Evidence: B.. After HT, children should be encouraged to increase

physical activity; daily intake of calcium with vitamin Dthrough diet or supplements should meet recommenda-tions for age.

Level of Evidence: C.. All adult HT recipients should begin anti-resorptive ther-

apy with bisphosphonates immediately after HT andcontinue it at least throughout the first post-operativeyear.

Level of Evidence: B.. Bisphosphonates can be used to treat bone loss in long-

term HT recipients and should be used in addition tocalcium and vitamin D.

Level of Evidence: C.0. In pediatric HT recipients who have not reached bone

maturity, bisphosphonates should be restricted to pa-tients with reduction in bone mass density associatedwith low-trauma fractures or vertebral compression.

Level of Evidence: B.

lass IIa:

. It is reasonable to perform spine radiographs in all adultHT candidates to detect existing fractures.

Level of Evidence: C.. After the first post-HT year, if glucocorticoids have been

discontinued and BMD is relatively normal (T score �

3

C

1

C

1

TT

RtC

1

2

3

4

5

6

7

8

9

C

1

C

1

RtC

1

2

939Costanzo et al. Guidelines for Heart Transplant Care

1.5), it is reasonable to stop bisphosphonates, whilemaintaining a high degree of vigilance for osteoporosis.

Level of Evidence: C.. Proximal femur and lumbar spine BMD should be as-

sessed by DEXA scanning in all adult patients 1 yearafter HT. Thereafter, annual reassessments are wise inpatients receiving CS and/or bisphosphonate therapy.However, it should be kept in mind that increases inBMD with bisphosphonates account for a small fractionof their efficacy in preventing bone fractures. It is rea-sonable to repeat BMD measurement in 2 years in pa-tients with osteopenia and in 3 years in patients withnormal bone density. Any clinical suggestion of fractureshould prompt bone radiographs.

Level of Evidence: C.

lass IIb:

. Active metabolites of vitamin D (calcidiol, alfacalcidol,and calcitriol) should not be regarded as the first-linetreatment for bone loss after HT. If they are used, fre-quent monitoring of urine and serum calcium levels isrequired, because hypercalcemia and hypercalciuria arecommon and may develop anytime during treatment.

Level of Evidence: B.

lass III:

. Calcitonin should not be used to prevent early bone lossafter HT.

Level of Evidence: B.

opic 10: Reproductive Health After Heartransplantation

ecommendations on Pregnancy After Heart Transplanta-ion:352–354

lass I:

. A multidisciplinary team, involving specialists in mater-nal and fetal medicine, cardiology and transplant medi-cine, anesthesia, neonatology, psychology, genetics, andsocial services, is important in the care of pregnant HTrecipients.

Level of Evidence: C.. The management plan for pregnant HT recipients should

be individualized according to the status of the motherand the allograft she received and is best achieved at theprimary transplant institution in collaboration with localor referring physicians.

Level of Evidence: C.. Individual factors in a HT recipient who wishes to be-

come pregnant should be considered, including the riskof acute rejection and infection, review of concomitanttherapy that is potentially toxic or teratogenic, and re-view of the adequacy of graft function. After carefulconsideration of these individual factors, patients shouldbe counseled on the risks of pregnancy and pregnancydiscouraged if graft dysfunction and significant CAV areexpected to preclude a successful outcome.

Level of Evidence: C.

. Pregnancy in a HT recipient should generally not beattempted sooner than 1 year after HT.

Level of Evidence: C.. In a HT recipient who wishes to become pregnant, base-

line tests should be obtained to determine the patient’scardiac status and should include an ECG and echocar-diogram (and coronary angiography if not performedwithin the previous 6 months) with the option of right-heart catheterization and EMB, if clinically indicated.

Level of Evidence: C.. Baseline assessment of renal and liver function should be

obtained in a pregnant HT recipient and frequent moni-toring of blood pressure, urine cultures, and surveillancefor pre-eclampsia and gestational diabetes should bedone.

Level of Evidence: C.. CNIs and CS should be continued in a pregnant HT

recipient, but MMF (class D) should be discontinued.Level of Evidence: C.

. Blood levels of CNI should be monitored closely duringpregnancy due to large fluctuations in levels during thepregnancy-related changes in plasma and interstitial vol-ume and hepatic and renal blood flow.

Level of Evidence: C.. Frequent surveillance for rejection is imperative in a

pregnant HT recipient, although surveillance EMB doneunder fluoroscopy should be avoided. An EMB underechocardiographic guidance or fluoroscopy with leadedpatient draping can be performed if necessary.

Level of Evidence: C.

lass IIb:

. The use of AZA (also class D), as a substitute for MMF,is somewhat controversial, and avoidance of both agentsin a pregnant HT recipient should be decided on the basisof the balance of maternal and fetal risk.

Level of Evidence: C.

lass III:

. It is uncertain whether the potential risks of drug expo-sure for the infant outweigh the benefits of breastfeeding,which is therefore not recommended for HT recipients.

Level of Evidence: C.

ecommendations for Contraception After Heart Transplan-ation:355,356

lass I:

. Before combination hormonal contraception is pre-scribed, a HT recipient should be screened for risk fac-tors for a hypercoagulable state (a strong family or per-sonal history of thromboembolic events).

Level of Evidence: C.. Combined hormonal contraception inhibits the CYP-450

3A4 pathway, and immunosuppressant drug blood levelsshould be monitored carefully when starting this therapyin HT recipients.

Level of Evidence: C.

3

C

1

C

1

2

RmC

1

2

3

4

RtC

1

2

3

TA

RAC

1

2

C

1

TH

Rg(

940 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

. Barrier methods provide inadequate pregnancy protec-tion and should be used as an adjunct to other methods inHT recipients. They should be recommended for allsexually active adolescents for sexually transmitted in-fection (STI) prevention.

Level of Evidence: B.

lass IIb:

. Intrauterine devices (IUD) have been generally not rec-ommended in HT recipients and, in particular, in nullip-arous patients because of the increased risk of IUDexpulsion in nulliparous women and because of concernsregarding increased risk of pelvic inflammatory infectionand infertility.

Level of Evidence: C.

lass III:

. Depo-medroxyprogesterone acetate has been associatedwith decreased bone density and, therefore, is not rou-tinely recommended for HT recipients.

Level of Evidence: C.. Hormonal contraception should not be prescribed in HT

recipients who have significant hypertension, knownCAV, estrogen-sensitive cancers, or active liver disease.

Level of Evidence: C.

ecommendations for the Management of Sexually Trans-itted Infections357:

lass I:

. Clinicians should obtain a confidential sexual historyfrom adolescent HT recipients and may consider routinereferral to an adolescent medicine specialist who willprovide thorough and confidential reproductive healthcare.

Level of Evidence: C.. Sexually active adolescents and adult HT recipients with

multiple partners should be advised to undergo screeningfor STI, including a complete anogenital examination toscreen for anogenital warts, molluscum, herpes simplexvirus (HSV), or other lesions at an appropriate clinic atregular intervals.

Level of Evidence: C.. A complaint of genitourinary symptoms or disclosure of

high-risk behavior should trigger a full evaluation forSTI in HT recipients. Genitourinary symptoms may alsobe an indication for empiric anti-microbial therapy whileawaiting results of STI screening.

Level of Evidence: C.. The quadrivalent human papillomavirus (HPV) vaccine

may prevent persistent HPV infection, cervical and vul-vovaginal cancer precursor lesions, and genital wartssecondary to HPV types 6, 11, 16, and 18. Womenshould receive all 3 doses before HT. There is no con-traindication to administering the vaccine to women afterHT, although no studies have confirmed immunogenicityor efficacy in this population.

Level of Evidence: C. C

ecommendations for the Management of Erectile Dysfunc-ion After Heart Transplantation358:lass I:

. Possible iatrogenic causes of erectile dysfunction (ED)should be identified in HT recipients, and alternativemedications should be used where possible.

Level of Evidence: C.. In HT recipients with ED, use of phosphodiesterase

inhibitors can be considered. Concomitant nitrate ther-apy is contraindicated similarly to the general popula-tion.

Level of Evidence: C.. In HT recipients with ED, consider referral to an ED

specialist for possible intra-cavernous injections of pros-taglandin E1 if phosphodiesterase inhibitors are ineffec-tive or contraindicated.

Level of Evidence: C.

opic 11: Exercise and Physical Rehabilitationfter Heart Transplantation

ecommendations for Exercise and Physical Rehabilitationfter Heart Transplantation:359–368

lass I:

. The routine use of cardiac rehabilitation with perfor-mance of aerobic exercise training is recommended afterHT. The short-term benefits of this approach includeimprovement in exercise capacity and possible modifi-cation of cardiovascular risk factors such as obesity,hypertension, and glucose intolerance. There is currentlyno information on potential long-term benefits.

Level of Evidence: B.. Resistance exercise is also strongly encouraged in HT

recipients to restore BMD and prevent the adverse ef-fects of CS and CNI therapy on skeletal muscle. Resis-tance exercise should be additive to other therapies forbone mineral loss and muscle atrophy.

Level of Evidence: B.

lass IIa:

. Exercise should be encouraged after pediatric HT, al-though no data on the long-term benefits exist. Exercisehas been shown to produce short-term improvements infunctional capacity and perhaps to decrease obesity-re-lated morbidity. Specific exercise programs should betailored to the specific needs and co-morbidities of theindividual HT recipient.

Level of Evidence: C.

opic 12: Management of Intercurrent Surgery ineart Transplant Recipients

ecommendations on the Management of Intercurrent Sur-ery in Heart Transplant Recipients369:See Table 15)

lass I:

1

2

3

4

5

6

TOT

RpC

1

2

3

4

5

6

941Costanzo et al. Guidelines for Heart Transplant Care

. HT recipients requiring intercurrent surgical proceduresshould have a full pre-operative assessment in collabo-ration with the transplant team, particularly in prepara-tion for major procedures requiring general or regionalanesthesia.

Level of Evidence: C.. For many surgical procedures, prophylactic anti-biotic

administration is now the norm. Protocols may needmodification in HT recipients. Aminoglycoside anti-bi-otics and erythromycin are best avoided because of therisk of worsening renal dysfunction when used in com-bination with CYA or TAC.

Level of Evidence: C.. When needed, blood products used in HT recipients

should be leukocyte poor. ABO-incompatible infant HTrecipients require specialized blood products and mustbe discussed with the transplant center.

Level of Evidence: C.. Anesthesia can be safely induced provided that there is

clear understanding that the HT is denervated. The rest-ing heart rate is usually higher in HT recipients. Al-though most allografts have a resting heart rate of ap-proximately 90 beats/min, some have resting sinus ratesas high as 130 beats/min, which do not require treatment.It must be remembered that a relative, symptomatic,bradycardia that requires treatment will not respond toatropine. Isoproterenol infusion and pacing are the usualmodes of management of HT bradyarrhythmias. Al-though uncommon, the likeliest sustained atrial arrhyth-mia is atrial flutter. Likewise, the denervated heart issuper-sensitive to adenosine, and the use of standarddoses to treat atrial tachyarrhythmias may result in pro-longed asystole. Amiodarone is recommended as thedrug of choice for atrial tachyarrhythmias in HT recipi-ents.

Level of Evidence: C.. Care with fluid balance is important because decreased

intravascular volume will exacerbate renal dysfunction,and fluid excess may not be well tolerated by HT recip-ients. For major surgery, CVP monitoring may be nec-essary.

Table 15 Conversion of Oral to Intravenous Doses ofImmunosuppressive Drugs

Cyclosporine One-third of oral daily doseeither as a continuousinfusion over 24 hours,or divided into two 6-hourly infusions twicedaily

Tacrolimus One-fifth of the oral dailydose as a continuousinfusion over 24 hours

Mycophenolatemofetil

Same as oral dose

Azathioprine Same as oral dose

Level of Evidence: C.

. Immunosuppression should not be discontinued or omit-ted without discussion with the HT team. However, itmay be prudent to omit the dose of CNI on the morningof surgery to avoid potentiating the detrimental effect ofdehydration on renal function. Thereafter, immunosup-pression should be continued as normal. If medicationscannot be given orally CYA should be given IV (often asa 6-hour infusion every 12 hours or as a continuousinfusion over 24 hours) at a third of the daily oral dose;TAC can be given IV at a dose one-fifth of the total dailyoral dose over 24 hours; AZA should be given IV oncedaily at the same dose as that taken orally; MMF can begiven IV at the same dose taken orally.

Level of Evidence: C.

opic 13: Return to Work or School andccupational Restrictions After Heartransplantation

ecommendations on Return to Work or School and Occu-ational Restrictions After Heart Transplantation:370–372

lass IIa:

. Health care providers should know that return to workfor HT recipients is possible, and not passively supportthe sick role of patients.

Level of Evidence: C.. Return to work should be discussed before HT as the

goal of post-operative rehabilitation, and not as an ex-ception.

Level of Evidence: C.. Patients should be encouraged to maintain their jobs as

long as possible before HT because this facilitates returnto work after HT.

Level of Evidence: C.. Short-term and long-term goals for returning to work

should be discussed as part of the discharge planningafter HT.

Level of Evidence: C.. An employment specialist (eg, a social worker) should

be appointed who can set up a proactive employmentatmosphere and facilitate the return to work process afterHT.

Level of Evidence: C.. This employment specialist should (1) perform a formal

assessment of the patient’s educational backgrounds,skills, beliefs, functional and physical limitations, andformer work experiences; (2) formulate a career planwith the patient that may help the patient to enter orrejoin the work force or acquire further vocational train-ing; (3) have knowledge of the job market and collabo-rate with the HT team in learning which physical limi-tations of the patient must be taken into account; (4)educate future employers about HT and share insightsabout an individual patient’s abilities and restrictions inview of post-operative rehabilitation.

Level of Evidence: C.

TH

RHC

1

2

3

4

5

6

T

RC

1

C

1

2

3

C

1

C

1

TT

RTC

1

TV

RCC

1

2

3

4

942 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

opic 14: Return to Operating a Vehicle Aftereart Transplantation

ecommendations for the Operation of a Vehicle Aftereart Transplantationlass I

. Assessment and discussion of the ability to drive a motorvehicle should be included in the early follow-up of HTrecipients.

Level of Evidence: C.. Gate stability, tremor, and other neurologic abnormali-

ties should be assessed before HT recipients obtain per-mission to drive.

Level of Evidence: C.. If symptomatic bradycardia is present after HT, the im-

plantation of a permanent pacemaker should be consid-ered before driving is permissible.

Level of Evidence: C.. The absence of severe hypoglycemic events should be

ascertained before HT recipients are permitted todrive.

Level of Evidence: C.. Occupational driving requires that HT recipients meet

their country’s requirements for occupational driving.Level of Evidence: C.

. A high level of scrutiny is required for HT recipientsrequesting to pilot an aircraft due to the risk of suddendeath associated with CAV.

Level of Evidence: C.

opic 15: Cardiac Retransplantation

ecommendations for Cardiac Retransplantation:373–375

lass I:

. Retransplantation is indicated in children with at leastmoderate systolic heart allograft dysfunction and/or se-vere diastolic dysfunction and at least moderate CAV.

Level of Evidence: B.

lass IIa:

. It is reasonable to consider listing for retransplantationthose adult HT recipients who develop severe CAV notamenable to medical or surgical therapy and symptomsof heart failure or ischemia.

Level of Evidence: C.. It is reasonable to consider listing for retransplantation

those HT recipients with heart allograft dysfunction andsymptomatic heart failure occurring in the absence ofacute rejection.

Level of Evidence: C.. It is reasonable to consider retransplantation in children

with normal heart allograft function and severe CAV.Level of Evidence: B.

lass IIb:

. Patients with severe CAV not amenable to medical orsurgical therapy with asymptomatic moderate to severeLV dysfunction may be considered for retransplantation.

Level of Evidence: C.

lass III:

. Adult and pediatric HT recipients with heart allograftfailure due to acute rejection or occurring less than 6months after the first HT and complicated by hemody-namic compromise are inappropriate candidates for re-transplantation.

Level of Evidence: C.

opic 16: Endocarditis Prophylaxis After Heartransplantation

ecommendations on Endocarditis Prophylaxis in Heartransplant Recipients:lass IIa:

. There are insufficient data to support specific recommen-dations for endocarditis prophylaxis in HT recipients.However, these patients are at risk of acquired valvulardysfunction, and the outcome of endocarditis is so poorin HT recipients that the use of anti-biotic prophylaxisfor dental procedures is considered reasonable in patientswith valvulopathies.

Level of Evidence: C.

opic 17: Frequency of Routine Tests and Clinicisits in Heart Transplant Recipients

ecommendation on the Frequency of Routine Tests andlinic Visits in Heart Transplant Recipients376:lass IIa:

. Lifelong follow-up by the transplant center is recom-mended for HT recipients due to (1) the possibility of acuteand/or chronic rejection; (2) the chronic use, toxicity, anddrug interactions of immunosuppressants and the associatedrisks for infection and malignancy; and (3) comorbiditiesrequiring specialized monitoring and management.

Level of Evidence: C.. Follow-up for HT recipients should be provided by a

multidisciplinary team, including surgeons, cardiolo-gists, nurses, psychologists, social workers, dieticians,and physiotherapists, among many others. Patients andcaregivers should recognize that HT requires a life-longcommitment to medical care.

Level of Evidence: C.. The frequency of follow-up visits for HT recipients will

depend on the time since HT and the post-operativeclinical course.

Level of Evidence: C.. In case of an uneventful recovery, follow-up visits are best

scheduled every 7 to 10 days during the first month afterHT, then every 14 days during the second month, monthlyduring the first year, and every 3 to 6 months thereafter.

Level of Evidence: C.

5

6

7

C

1

2

F

3

TtT

RpC

1

2

3

4

5

6

7

8

BBBBS

943Costanzo et al. Guidelines for Heart Transplant Care

. The frequency of follow-up should be increased if com-plications occur, particularly in patients with challengingmedical or psychosocial conditions.

Level of Evidence: C.. Ancillary services, including home care nursing, cardiac

rehabilitation, psychologic support, nutritional planning,or patient support groups may also be used as resourcesin the follow-up of HT recipients, with the understandingthat providers of community health care services mustcommunicate with the clinicians at the transplant centerto ensure that the care delivered complies with the HTcenter’s standards.

Level of Evidence: C.. Local health professionals should inform the transplant

center in the case of the following events: (1) hospital-ization for any reason; (2) change in medication, includ-ing the addition of any anti-biotic, anti-fungal, or anti-viral therapy for confirmed or presumed infection; (3)hypotension or unexplained drop in systolic blood pres-sure � 20 mm Hg from baseline; (4) increase in restingheart rate � 10 beats/min over baseline; (5) fever �101°F (38°C) or any unexplained fever � 100.5° F for �48 hours (38°C); (6) � 2-pound weight gain in 1 week(ie, 900 g or more); (7) unexplained weight loss of � 5pounds (ie, 2.3 kg); (8) elective surgery; (9) increasedshortness of breath; (10) pneumonia or any respiratoryinfection; (11) syncope; (12) chest pain other than mus-culoskeletal symptoms; (13) decline � 10% in forcedexpiratory volume in 1 second; (14) abdominal pain;(15) nausea, vomiting or diarrhea; (16) cerebral vascularevent, seizure, or mental status changes.

Level of Evidence: C.

lass I:

. In addition to routine outpatient follow-up visits, HTrecipients should have more prolonged visits every 1 to2 years for more detailed clinical assessment.

Level of Evidence: B.. The purpose of the follow-up visits is to monitor for

rejection and screen for adverse events, and may includethe following: (1) a complete physical examination; (2)review of the medication and changes to the medicationbased on the results of the examinations; (3) blood work;(4) echocardiogram; (5) coronary angiography and IVUS(every 1 to 2 years); (6) EMB according to the typicalschedule outlined in the chart below; (7) additional ed-ucation and/or interaction with members of the multidis-ciplinary team. An example of a typical biopsy schedulefor the first year could be:

iopsy 1, 2, 3, 4, and 5: Weeklyiopsy 6, 7, and 8: Every 14 daysiopsy 9 and 10: Every 3 weeksiopsy 11, 12, and 13: Every 4 weeksubsequent biopsies duringthe 1st year after HT: Every 5 to 6 weeks

This recommendation is addressed in more detail in Taskorce 2.

Level of Evidence: B.

. In pediatric practice, far fewer biopsies are performeddue to the need for general anesthesia in small childrenand the difficulties with venous access and bioptomemanipulation in small hearts and vessels. There is noconsensus on the frequency of biopsy in children. Somecenters do no EMB at all, but instead use detailed echo-cardiographic assessment. Besides scheduled clinic ap-pointments, the patients should be encouraged to contactthe transplant center with questions, concerns, or unex-pected symptoms.

Level of Evidence: C.

opic 18: Psychologic Issues Particularly Relatedo Adherence to Medical Therapy in Heartransplant Recipients

ecommendations on Psychologic Issues After Heart Trans-lantation:377–383

lass IIa:

. Adherence with the prescribed regimen should be rou-tinely assessed at every HT outpatient clinic visit.

Level of Evidence: C.. Because there is currently no gold standard for adherence

assessment in HT recipients, it is recommended to com-bine methods to increase accuracy of assessment (eg, acombination of self-report or parent report in case ofchildren, drug levels assessment, and clinical judgment).

Level of Evidence: C.. Attention should be given not only to adherence to the

immunosuppressive regimen but also to all other healthrecommendations appropriate for HT recipients.

Level of Evidence: C.. Barriers to adherence should be discussed in an open,

non-threatening way during visits with HT recipients.Level of Evidence: C.

. Tailored interventions, in close collaboration with theHT recipient and his or her family, should be consideredand their efficacy explored. Strategies that seem mosteffective include offering education repeatedly, reducingthe complexity of the medication regimen, providingfeedback on a patient’s behavior, and combining strate-gies.

Level of Evidence: C.. Strategies to enhance maturity and independence may be

particularly helpful in the adolescent HT recipients.Level of Evidence: C.

. Because adherence to medical recommendations is acomplex issue, health care teams would benefit fromtraining in measuring adherence, discussing its barriers,and implementing adherence-enhancing interventionsfor HT recipients.

Level of Evidence: C.. Each HT center should closely collaborate with a spe-

cialized nurse or psychologist who can screen and mon-

9

1

C

1

2

3

TPT

Rf3

C

1

2

3

TT

RTC

1

2

3

AA

944 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

itor all HT recipients at risk for non-adherence. Investingin specialized staff may result in better transplant out-comes in the long-term, although further studies testingthe efficacy of adherence-enhancing interventions arewarranted.

Level of Evidence: C.. Depressive symptoms should be regularly evaluated dur-

ing follow-up of HT recipients. This can best be done byuser-friendly, validated screening instruments. All pa-tients with elevated scores should be referred to special-ized treatment.

Level of Evidence: C.0. Each HT team should include a psychologist who is

qualified to detect and treat depression. Multidisci-plinary treatment teams are better prepared to addresspsychosocial risk factors for poor outcomes after HT.

Level of Evidence: C.

lass I:

. Serotonin reuptake inhibitors, particularly citalopram,and new-generation anti-depressants (mirtazapine) maybe the best choice for HT recipients because they have nosignificant impact on blood pressure, heart rate, rhythm,or conduction intervals.

Level of Evidence: B.. Agents that interact with the metabolism of CYA and

TAC via the CYP450 system (eg, fluvoxamine, nefaz-odone) should be avoided because they may alter CNIlevels.

Level of Evidence: B.. Tricyclic anti-depressants (eg, imipramine, desipramine,

amitriptyline, and clomipramine) are associated with car-diovascular toxicity (conduction delay, orthostatic hypo-tension, and anti-cholinergic effects) and may lower sei-zure thresholds, and therefore, their use should berestricted to HT recipients with severe depression refrac-tory to other therapies. Monoamine oxidase inhibitors(MAOIs) should be avoided because of their hypotensiveeffects, interactions with anesthetic and pressor agents,and need for dietary restrictions. Herbal medicines suchas St. John’s wort (Hypericum perforatum) can be harm-ful because it lowers CYA levels.

Level of Evidence: B.

opic 19: Management of the Transition fromediatric to Adult Care After Heartransplantation

ecommendations on the Management of the Transitionrom Pediatric to Adult Care After Heart Transplantation:84–386

lass I:

. Critical milestones to be achieved by pediatric HT re-cipients before transition to adult care include (1) under-standing of and ability to describe the original cause oftheir organ failure and need for HT (initial education

may have been primarily provided to the parents of the A

HT recipient, and repetition is necessary to ensure un-derstanding of the clinical condition by the HT recipient;(2) awareness of the long and short-term clinical impli-cations of chronic immunosuppression (infection pre-vention, cancer surveillance, academic and vocationalaspirations); (3) comprehension of the impact of HTstatus on sexuality and reproductive health (impact ofpregnancy, effect of medications on fertility, any poten-tial teratogenicity of medications, role of genetic coun-seling and genetic risk of disease recurrence in offspring,and increased susceptibility to sexually transmitted dis-ease); (4) demonstration of a sense of responsibility forself-care (knowledge of medications, ability to obtainprescription refills, adherence to medication and officevisits schedules, ability to independently communicatewith health providers, recognition of symptoms andsigns requiring immediate medical attention, and under-standing of health care coverage and eligibility require-ments).

Level of Evidence: C.. Health care providers should simultaneously prepare the

parents for the transition from pediatric to adult care byencouraging independence and self-responsibility in thechild.

Level of Evidence: C.. Practitioners who care for adults should cultivate part-

nerships with their pediatric colleagues to gain insightinto the care of adolescents and the impact of childhoodchronic disease on development and management ofchildhood causes of end-stage organ failure and congen-ital diseases. Ideal adult site resources also include adedicated transfer liaison nurse coordinator, a socialworker, and a reproductive specialist.

Level of Evidence: C.

opic 20: Principles of Shared Care After Heartransplantation

ecommendations on Principles of Shared Care After Heartransplantation:lass I:

. The HT team should ensure that other involved physiciansknow telephone numbers and electronic mail addresses ofthe HT team to enable contact at all times and guaranteeprompt responses to referring physicians’ queries.

Level of Evidence: C.. It is helpful for physicians outside the HT team to receive

the patient’s plan for scheduled HT office visits at thetransplant center.

Level of Evidence: C.. Formal procedures should be instituted to regularly in-

form the referring physician of clinical results and med-ical regimens.

Level of Evidence: C.

BBREVIATIONSAIR � atrium paced, atrium sensed inhibited rate modulation

CC � American College of Cardiology

AAAAAAaAAABBBCCCCCCCCCCCCCCCDmDDEEEEEEEFGHHHHHHHHII

IIITIIILLLMMMMMmMPPPPPPPPPPPRrRsSSSTTTTTVVV

CC

mti

945Costanzo et al. Guidelines for Heart Transplant Care

CEI � angiotensin converting enzyme inhibitorCT � activated clotting timeDA � American Diabetes AssociationHA � American Heart AssociationMR � antibody-mediated rejectionP � aerosolized pentamidine

PTT � activated partial thromboplastin timeRB � angiotensin receptor blockerTG � anti-thymocyte globulinZA � azathioprineiV � biventricularMD � bone mass densityNP � brain natriuretic peptideAV � cardiac allograft vasculopathyCB � calcium channel blockerEDIA � cloned enzyme donor immunoassay methodI � cardiac indexKD � chronic kidney diseaseO � cardiac outputPB � cardiopulmonary bypassMV � cytomegalovirusNI � calcineurin inhibitorRP � C-reactive proteinS � corticosteroidT � computed tomographyVP � central venous pressureYA � cyclosporineYP3A � cytochrome P-450 3A4DDR � dual-paced, dual-sensed, dual-response to sensing, rateodulationEXA � dual energy x-ray absorptiometrySA � donor specific antibodyCG � electrocardiogramD � erectile dysfunctionCMO � extracorporeal membrane oxygenationMB � endomyocardial biopsyMIT � enzyme multiplied immunoassay techniqueSC � European Society of CardiologyVL � everolimusFP � fresh frozen plasmaFR � glomerular filtration rategb � hemoglobinIT � heparin-induced thrombocytopeniaLA � human leukocyte antigenPLC � high-performance liquid chromatographyPV � human papillomavirusRS � Heart Rhythm SocietySV � herpes simplex virusT � heart transplant

CU � intensive care unitg � immunoglobulin

Name Commercial Int

Allen S. Anderson XDxDavid A. Baran XDx

AstellasGileadGSKUnited Therape

Geetha Bhat Nothing to dis

gG � immunoglobulin GNR � international normalized unitSHLT � International Society for Heart and Lungransplantation

UD � intrauterine deviceV � intravenousVUS � intravascular ultrasoundV � left ventricleVEF � left ventricular ejection fractionVH � left ventricular hypertrophyAOI � monoamine oxidase inhibitorsCS � mechanical circulatory supportDRD equation � modified diet in renal disease equationMF � mycophenolate mofetilPA � mycophenolic acidTOR � mammalian target of rapamycinVO2 � mixed venous oxygen

AWP � pulmonary artery wedge pressureCC � prothrombin plasma concentratesFA-100 � platelets function assay 100GF � primary graft failureRA � panel reactive antibodiesRES � posterior reversible leukoencephalopathySI � proliferation signal inhibitorTLD � posttransplant lymphoproliferative disorderT � prothrombin timeTT � partial thromboplastin timeVR � pulmonary vascular resistanceAP � right atrial pressure

FVII � recombinant factor 7V � right ventricle

Cr � serum creatinineRL � sirolimusTI � sexually transmitted infectionVT � sustained ventricular tachycardiaAC � tacrolimusEE � transesophageal echocardiogramMP/SMZ � trimethoprim/sulfamethoxazoleTE � transthoracic echocardiogramV � tricuspid valveAD � ventricular assist deviceER � ventricular evoked responsesT � ventricular tachycardia

onflict of Interest Disclosuresontributing Writers and ReviewsEach writing group member and editorial oversight com-

ittee member completed a disclosure form that is main-ained on file at the ISHLT headquarters. The conflict ofnterest disclosures are as follows:

Relationship

Grant research/supportGrant research/supportGrant research/supportSpeakers bureauSpeakers bureauAdvisory board

erest

uticsclose

946 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

Name Commercial Interest Relationship

Michael Burch Novartis ConsultantCharles Canter Novartis Consultant

Blue Cross/Blue Shield consultantMichael Carboni Nothing to discloseMichael Chan Nothing to discloseRichard Chinnock Nothing to discloseMaria Rosa Costanzo CHF Solutions Consultant; speaker

St. Jude Speaker; research grantCardiomems Research grantParacor Research grant

Marisa Crespo-Leiro Roche Research grant/speakerNovartis Research grant/speakerWyeth Research grantAstellas Pharma Research grant/speaker

Diego Delgado Astellas Research grantNovartis Research grant

Reynolds Delgado Gilead Speakers bureauEli Lily Speakers bureau

Thomas Dengler Nothing to discloseShashank Desai Nothing to discloseAnne Dipchand Nothing to discloseFabienne Dobbels Astellas Speaker/research grant

BMS Scientific advisor/research grantSpeakers bureau

Novartis Scientific advisor/speakerHoward Eisen Novartis Grant/research support

Novartis Consultant/scientific advisorWyeth Grant/research supportWyeth Scientific medical/advisory

Savitri Fedson Nothing to discloseDavid Feldman Nothing to disclosePatrick Fis her Nothing to discloseMaria Frigerio Novartis Speaker

Genzyme SpeakerLee Goldberg Thoratec Consultant

Novartis Research grantAlere Sci/medical advisory boardMedtronic ConsultantGSK Speakers bureau

Gonzalo Gonzalez-Stawinski Thoratec Honorarium, travel grantKathleen Grady Nothing to discloseJeff Hosenpud Nothing to discloseSharon Hunt Nothing to discloseMaryl Johnson CVS (Caremark) ConsultantWalter Kao Nothing to discloseAnne Keogh Pfizer Research grant

Heartware Research grantNovartis Research grantBayer Research grantBayer Sci/medical advisory boardGSK Research grantGSK Sci/medical advisory boardActelion Research grantActelion Sci/medical advisory boardActelion Speakers bureau

Abdallah Kfoury XDx Scientific medical/advisoryXDx Grant research/supportNovartis Research grant

Daniel Kim Nothing to discloseJon Kobashigawa Novartis Research grants

R

947Costanzo et al. Guidelines for Heart Transplant Care

eferences

1. Young JB, Hauptman PJ, Naftel DC, et al. Determinants of early graftfailure following cardiac transplantation, a 10-year, multi-institutional,multivariable analysis. J Heart Lung Transplant 2001;20:212.

2. Lietz K, John R, Mancini DM, Edwards NM. Outcomes in cardiactransplant recipients using allografts from older donors versus mor-tality on the transplant waiting list; implications for donor selectioncriteria. J Am Coll Cardiol 2004;43:1553-61.

3. Kubak BM, Gregson AL, Pegues DA, et al. Use of hearts trans-planted from donors with severe sepsis and infectious deaths. J HeartLung Transplant 2009;28:260-5.

4. Brieke A, Krishnamani R, Rocha MJ, et al. Influence of donor co-caine use on outcome after cardiac transplantation: analysis of theUnited Network for Organ Sharing Thoracic Registry. J Heart Lung

Name Commercial Inte

NovartisXDxGenzyme

Jacqueline Lamour Nothing to disclClive Lewis Roche

ActelionLuigi Martinelli Nothing to disclDavid McGiffin Nothing to disclBruno Meiser AstellasJohn O’Connell XDXGareth Parry Nothing to disclJignesh Patel Novartis

QRS Medical SysDaniela Pini Nothing to disclJoseph Rogers ThoratecHeather Ross Wyeth

NovartisStuart Russell ThoratecMichael Shullo Nothing to disclJon Smith Nothing to disclRandall Starling Novartis

NovartisMedtronicMedtronicXDx

Josef Stehlik Nothing to disclDavid Taylor Nothing to disclJeffrey Teuteberg Thoratec Corpora

XDx

Jeffrey Towbin PGx HealthGeneD

Patricia Uber Geron CorpSteven Webber Nothing to disclGene Wolfel Nothing to disclAndreas Zuckermann Novartis

NovartisAstellasWyethRocheGenzymeGenzyme

Transplant 2008;27:1350-2.

5. De La Zerda DJ, Cohen O, Beygui RE, et al. Alcohol use in donors isa protective factor on recipients’ outcome after heart transplantation.Transplantation 2007;83:1214-8.

6. Smith JA, Bergin PJ, Williams TJ, Esmore DS. Successful hearttransplantation with cardiac allografts exposed to carbon monoxidepoisoning. J Heart Lung Transplant 1992;11:698-700.

7. Snyder JW, Unkle DW, Nathan HM, Yang SL. Successful donationand transplantation of multiple organs from a victim of cyanidepoisoning. Transplantation 1993;55:425-7.

8. Navia JL, Atik FA, Marullo A, et al. Bench repair of donor aorticvalve with minimal access orthotopic heart transplantation. Ann Tho-rac Surg 2005;80:313-5.

9. Laks H, Scholl FG, Drinkwater DC, et al. The alternate recipient list for hearttransplantation: does it work? J Heart Lung Transplant 1997;16:735-42.

10. Young JB, Naftel DC, Bourge RC, et al. Matching the heart donor and

Relationship

Scientific medical/advisoryResearch grantResearch grant

Travel grantTravel grants

Consultant/scientific advisorHonorarium

Grant research/supportSci/med advisory board

Consultant/scientific advisorDSMB MemberPrincipal investigatorConsultant

Grant/research supportConsultant/scientific advisorGrant/research supportConsultant/scientific advisorGrant research/support

ConsultantConsultantGrant research/supportSpeakerConsultant/scientific advisorSpeaker’s bureauConsultant

Speakers bureauConsultant/scientific advisorSpeaker’s bureauGrant/research supportResearch grantSpeakers bureauGrant/research support

rest

ose

oseose

ose

t.ose

oseose

oseosetion

oseose

heart transplant recipient. Clues for successful expansion of the donor

948 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

pool: a multivariable, multiinstitutional report. The Cardiac TransplantResearch Database Group. J Heart Lung Transplant 1994;13:353-64.

11. Blackbourne LH, Tribble CG, Langenburg SE, et al. Successful useof undersized donors for orthotopic heart transplantation—with acaveat. Ann Thorac Surg 1994;57:1472-5.

12. Sethi GK, Lanauze P, Rosado LJ, et al. Clinical significance ofweight difference between donor and recipient in heart transplanta-tion. J Thorac Cardiovasc Surg 1993;106:444-8.

13. Bull DA, Stahl RD, McMahan DL, et al. The high risk heart donor:potential pitfalls. J Heart Lung Transplant 1995;14:424-8.

14. Mackintosh AF, Carmichael DJ, Wren C, Cory-Pearce R, En-glish TA. Sinus node function in first three weeks after cardiactransplantation. Br Heart J 1982;48:584-8.

15. Leonelli FM, Pacifico A, Young JB. Frequency and significance ofconduction defects early after orthotopic heart transplantation. Am JCardiol 1994;73:175-9.

16. Leeman M, Van CM, Vachiery JL, Antoine M, Leclerc JL. Determi-nants of right ventricular failure after heart transplantation. ActaCardiol 1996;51:441-9.

17. Kimball TR, Witt SA, Daniels SR, Khoury PR, Meyer RA. Fre-quency and significance of left ventricular thickening in transplantedhearts in children. Am J Cardiol 1996;77:77-80.

18. Mahle WT, Cardis BM, Ketchum D, et al. Reduction in initial ven-tricular systolic and diastolic velocities after heart transplantation inchildren: improvement over time identified by tissue Doppler imag-ing. J Heart Lung Transplant 2006;25:1290-6.

19. Asante-Korang A, Fickey M, Boucek MM, Boucek RJ Jr. Diastolicperformance assessed by tissue Doppler after pediatric heart trans-plantation. J Heart Lung Transplant 2004;23:865-72.

20. Stinson EB, Caves PK, Griepp RB, et al. Hemodynamic observationsin the early period after human heart transplantation. J Thorac Car-diovasc Surg 1975;69:264-70.

21. Rothman SA, Jeevanandam V, Combs WG, et al. Eliminating brady-arrhythmias after orthotopic heart transplantation. Circulation 1996;94(9 suppl):II278-82.

22. Zieroth S, Ross H, Rao V, et al. Permanent pacing after cardiactransplantation in the era of extended donors. J Heart Lung Trans-plant 2006;25:1142-7.

23. Costanzo-Nordin MR, Liao YL, Grusk BB, et al. Oversizing of donorhearts: beneficial or detrimental? J Heart Lung Transplant 1991;10:717-30.

24. Minev PA, El-Banayosy A, Minami K, et al. Differential indicationfor mechanical circulatory support following heart transplantation.Intensive Care Med 2001;27:1321-7.

25. Ibrahim M, Hendry P, Masters R, et al. Management of acute severeperioperative failure of cardiac allografts: a single-centre experiencewith a review of the literature. Can J Cardiol 2007;23:363-7.

26. Santise G, Petrou M, Pepper JR, et al. Levitronix as a short-termsalvage treatment for primary graft failure after heart transplantation.J Heart Lung Transplant 2006;25:495-8.

27. Huang J, Trinkaus K, Huddleston CB, et al. Risk factors for primarygraft failure after pediatric cardiac transplantation: importance ofrecipient and donor characteristics. J Heart Lung Transplant 2004;23:716-22.

28. Mehra MR, Kobashigawa J, Starling R, et al. Listing criteria for hearttransplantation: International Society for Heart and Lung Transplan-tation guidelines for the care of cardiac transplant candidates—2006.J Heart Lung Transplant 2006;25:1024–2.

29. Bhatia SJ, Kirshenbaum JM, Shemin RJ, et al. Time course of reso-lution of pulmonary hypertension and right ventricular remodelingafter orthotopic cardiac transplantation. Circulation 1987;76:819-26.

30. Stobierska-Dzierzek B, Awad H, Michler RE. The evolving manage-ment of acute right-sided heart failure in cardiac transplant recipients.J Am Coll Cardiol 2001;38:923-31.

31. Kieler-Jensen N, Lundin S, Ricksten SE. Vasodilator therapy afterheart transplantation: effects of inhaled nitric oxide and intravenousprostacyclin, prostaglandin E1, and sodium nitroprusside. J Heart

Lung Transplant 1995;14:436-43.

32. Jeevanandam V, Russell H, Mather P, et al. Donor tricuspid annulo-plasty during orthotopic heart transplantation: long-term results of aprospective controlled study. Ann Thorac Surg 2006;82:2089-95.

33. Wong RC, Abrahams Z, Hanna M, et al. Tricuspid regurgitation aftercardiac transplantation: an old problem revisited. J Heart Lung Trans-plant 2008;27:247-52.

34. Hauptman PJ, Couper GS, Aranki SF, et al. Pericardial effusions af-ter cardiac transplantation. J Am Coll Cardiol 1994;23:1625-9.

35. Quin JA, Tauriainen MP, Huber LM, et al. Predictors of pericardialeffusion after orthotopic heart transplantation. J Thorac CardiovascSurg 2002;124:979-83.

36. Chen EP, Bittner HB, Davis RD, Van TP. Hemodynamic and inotro-pic effects of milrinone after heart transplantation in the setting ofrecipient pulmonary hypertension. J Heart Lung Transplant 1998;17:669-78.

37. Leyh RG, Kofidis T, Struber M, et al. Methylene blue: the drug ofchoice for catecholamine-refractory vasoplegia after cardiopulmo-nary bypass? J Thorac Cardiovasc Surg 2003;125:1426-31.

38. Argenziano M, Choudhri AF, Oz MC, et al. A prospective random-ized trial of arginine vasopressin in the treatment of vasodilatoryshock after left ventricular assist device placement. Circulation 1997;96(9 suppl):II-90.

39. Morales DL, Garrido MJ, Madigan JD, et al. A double-blind random-ized trial: prophylactic vasopressin reduces hypotension after cardio-pulmonary bypass. Ann Thorac Surg 2003;75:926-30.

40. Armitage JM, Hardesty RL, Griffith BP. Prostaglandin E1: an effec-tive treatment of right heart failure after orthotopic heart transplan-tation. J Heart Transplant 1987;6:348-51.

41. Pascual JM, Fiorelli AI, Bellotti GM, Stolf NA, Jatene AD. Prosta-cyclin in the management of pulmonary hypertension after hearttransplantation. J Heart Transplant 1990;9:644-51.

42. Theodoraki K, Tsiapras D, Tsourelis L, et al. Inhaled iloprost in eightheart transplant recipients presenting with post-bypass acute rightventricular dysfunction. Acta Anaesthesiol Scand 2006;50:1213-7.

43. Ardehali A, Hughes K, Sadeghi A, et al. Inhaled nitric oxide forpulmonary hypertension after heart transplantation. Transplanta-tion 2001;72:638-41.

44. Auler Junior JO, Carmona MJ, Bocchi EA, et al. Low doses of in-haled nitric oxide in heart transplant recipients. J Heart Lung Trans-plant 1996;15:443-50.

45. De Santo LS, Mastroianni C, Romano G, et al. Role of sildenafil inacute posttransplant right ventricular dysfunction: successful experi-ence in 13 consecutive patients. Transplant Proc 2008;40:2015-8.

46. Kirklin JK, Young JB, McGiffin DC. Heart transplantation. Philadel-phia: Churchill Livingstone, 2002.

47. Haddad F, Hunt SA, Rosenthal DN, Murphy DJ. Right ventricularfunction in cardiovascular disease, part I: anatomy, physiology, ag-ing, and functional assessment of the right ventricle. Circula-tion 2008;117:1436-48.

48. Arafa OE, Geiran OR, Andersen K, et al. Intraaortic balloon pumpingfor predominantly right ventricular failure after heart transplantation.Ann Thorac Surg 2000;70:1587-93.

49. Jurmann MJ, Wahlers T, Coppola R, Fieguth HG, Haverich A. Earlygraft failure after heart transplantation: management by extracorpo-real circulatory assist and retransplantation. J Heart Transplant 1989;8:474-8.

50. Tenderich G, Koerner MM, Stuettgen B, et al. Mechanical circula-tory support after orthotopic heart transplantation. Int J Artif Or-gans 1998;21:414-6.

51. Barnard SP, Hasan A, Forty J, Hilton CJ, Dark JH. Mechanical ven-tricular assistance for the failing right ventricle after cardiac trans-plantation. Eur J Cardiothorac Surg 1995;9:297-9.

52. Chen JM, Levin HR, Rose EA, et al. Experience with right ventric-ular assist devices for perioperative right-sided circulatory failure.Ann Thorac Surg 1996;61:305-10.

53. Costard-Jackle A, Fowler MB. Influence of preoperative pulmonaryartery pressure on mortality after heart transplantation: testing of poten-tial reversibility of pulmonary hypertension with nitroprusside is useful

in defining a high risk group. J Am Coll Cardiol 1992;19:48-54.

949Costanzo et al. Guidelines for Heart Transplant Care

54. Bourge RC, Naftel DC, Costanzo-Nordin MR, et al. Pretransplanta-tion risk factors for death after heart transplantation: a multiinstitu-tional study. The Transplant Cardiologists Research Database Group.J Heart Lung Transplant 1993;12:549-62.

55. Chen JM, Levin HR, Michler RE, et al. Reevaluating the significanceof pulmonary hypertension before cardiac transplantation: determi-nation of optimal thresholds and quantification of the effect of re-versibility on perioperative mortality. J Thorac CardiovascSurg 1997;114:627-34.

56. Erickson KW, Costanzo-Nordin MR, O’Sullivan EJ, et al. Influenceof preoperative transpulmonary gradient on late mortality after or-thotopic heart transplantation. J Heart Transplant 1990;9:526-37.

57. Bando K, Konishi H, Komatsu K, et al. Improved survival followingpediatric cardiac transplantation in high-risk patients. Circula-tion 1993;88:II218-23.

58. Bauer J, Dapper F, Demirakca S, et al. Perioperative management ofpulmonary hypertension after heart transplantation in childhood.J Heart Lung Transplant 1997;16:1238-47.

59. Addonizio LJ, Gersony WM, Robbins RC, et al. Elevated pulmonaryvascular resistance and cardiac transplantation. Circulation 1987;76:V52-5.

60. Bailey LL, Gundry SR, Razzouk AJ, et al. Bless the babies: onehundred fifteen late survivors of heart transplantation during the firstyear of life. The Loma Linda University Pediatric Heart TransplantGroup. J Thorac Cardiovasc Surg 1993;105:805-14.

61. Stecker EC, Strelich KR, Chugh SS, Crispell K, McAnulty JH. Ar-rhythmias after orthotopic heart transplantation. J Card Fail 2005;11:464-72.

62. Gao SZ, Hunt SA, Wiederhold V, Schroeder JS. Characteristics ofserial electrocardiograms in heart transplant recipients. AmHeart J 1991;122:771-4.

63. Holt ND, McComb JM. Cardiac transplantation and pacemakers:when and what to implant. Card Electrophysiol Rev 2002;6:140-51.

64. Miyamoto Y, Curtiss EI, Kormos RL, et al. Bradyarrhythmia afterheart transplantation. Incidence, time course, and outcome. Circula-tion 1990;82(5 Suppl):IV313-7.

65. Pavri BB, O’Nunain SS, Newell JB, Ruskin JN, William G. Preva-lence and prognostic significance of atrial arrhythmias after ortho-topic cardiac transplantation. J Am Coll Cardiol 1995;25:1673-80.

66. Vaseghi M, Boyle NG, Kedia R, et al. Supraventricular tachycardiaafter orthotopic cardiac transplantation. J Am Coll Cardiol 2008;51:2241-9.

67. Ellenbogen KA, Thames MD, DiMarco JP, Sheehan H, Lerman BB.Electrophysiological effects of adenosine in the transplanted humanheart. Evidence of supersensitivity. Circulation 1990;81:821-8.

68. Rubel JR, Milford EL, McKay DB, Jarcho JA. Renal insufficiencyand end-stage renal disease in the heart transplant population. J HeartLung Transplant 2004;23:289-300.

69. Costanzo MR, Guglin ME, Saltzberg MT, et al. Ultrafiltration versusintravenous diuretics for patients hospitalized for acute decompen-sated heart failure. J Am Coll Cardiol 2007;49:675-83.

70. Boyle JM, Moualla S, Arrigain S, et al. Risks and outcomes of acutekidney injury requiring dialysis after cardiac transplantation. Am JKidney Dis 2006;48:787-96.

71. Anselm A, Cantarovich M, Davies R, Grenon J, Haddad H. Pro-longed basiliximab use as an alternative to calcineurin inhibition toallow renal recovery late after heart transplantation. J Heart LungTransplant 2008;27:1043-5.

72. Furnary AP, Gao G, Grunkemeier GL, et al. Continuous insulin in-fusion reduces mortality in patients with diabetes undergoing coro-nary artery bypass grafting. J Thorac Cardiovasc Surg 2003;125:1007-21.

73. Wilson M, Weinreb J, Hoo GW. Intensive insulin therapy in criticalcare: a review of 12 protocols. Diabetes Care 2007;30:1005-11.

74. Hawkins JA, Breinholt JP, Lambert LM, et al. Class I and class IIanti-HLA antibodies after implantation of cryopreserved allograftmaterial in pediatric patients. J Thorac Cardiovasc Surg 2000;119:324-30.

75. Pagani FD, Dyke DB, Wright S, Cody R, Aaronson KD. Develop-

ment of anti-major histocompatibility complex class I or II antibodies

following left ventricular assist device implantation: effects onsubsequent allograft rejection and survival. J Heart LungTransplant 2001;20:646-53.

76. Feingold B, Bowman P, Zeevi A, et al. Survival in allosensitizedchildren after listing for cardiac transplantation. J Heart Lung Trans-plant 2007;26:565–1.

77. Pollack-BarZiv SM, den Hollander N, Ngan B-Y, et al. Pediatricheart transplantation in HLA-sensitized patients: evolving manage-ment and assessment of intermediate-term outcomes in a high-riskpopulation. Circulation 2007;116(11 suppl):172-8.

78. Betkowski AS, Graff R, Chen JJ, Hauptman PJ. Panel-reactive anti-body screening practices prior to heart transplantation. J Heart LungTransplant 2002;21:644-50.

79. Tambur AR, Pamboukian SV, Costanzo MR, et al. The presence ofHLA-directed antibodies after heart transplantation is associated withpoor allograft outcome. Transplantation 2005;80:1019-25.

80. Kobashigawa J, Mehra M, West L, et al. Report from a consensusconference on the sensitized patient awaiting heart transplantation.J Heart Lung Transplant 2009;28:213-25.

81. Gebel HM, Bray RA, Nickerson P. Pre-transplant assessment of do-nor-reactive, HLA-specific antibodies in renal transplantation: con-traindication vs. risk. Am J Transplant 2003;3:1488-500.

82. Zangwill S, Ellis T, Stendahl G, et al. Practical application of thevirtual crossmatch. Pediatr Transplant 2007;11:650-4.

83. Pei R, Lee JH, Shih NJ, Chen M, Terasaki PI. Single human leuko-cyte antigen flow cytometry beads for accurate identification of hu-man leukocyte antigen antibody specificities. Transplantation 2003;75:43-9.

84. Vaidya S. Clinical importance of anti-human leukocyte antigen-spe-cific antibody concentration in performing calculated panel reactiveantibody and virtual crossmatches. Transplantation 2008;85:1046-50.

85. Takemoto SK, Zeevi A, Feng S, et al. National conference to assessantibody-mediated rejection in solid organ transplantation. Am JTransplant 2004;4:1033-41.

86. Pisani BA, Mullen GM, Malinowska K, et al. Plasmapheresis withintravenous immunoglobulin G is effective in patients with elevatedpanel reactive antibody prior to cardiac transplantation. J Heart LungTransplant 1999;18:701-6.

87. Holt DB, Lublin DM, Phelan DL, et al. Mortality and morbidity inpre-sensitized pediatric heart transplant recipients with a positivedonor crossmatch utilizing peri-operative plasmapheresis and cyto-lytic therapy. J Heart Lung Transplant 2007;26:876-82.

88. West LJ, Pollock-Barziv SM, Dipchand AI, et al. ABO-incompatibleheart transplantation in infants. N Engl J Med 2001;344:793-800.

89. Roche SL, Burch M, O’Sullivan J, et al. Multicenter experience ofABO-incompatible pediatric cardiac transplantation. Am J Trans-plant 2008;8:208-15.

90. West LJ, Karamlou T, Dipchand AI, et al. Impact on outcomes afterlisting and transplantation, of a strategy to accept ABO blood group-incompatible donor hearts for neonates and infants. J Thorac Cardio-vasc Surg 2006;131:455-61.

91. Dipchand AI, Benson L, McCrindle BW, Coles J, West L. Mycophe-nolate mofetil in pediatric heart transplant recipients: a single-centerexperience. Pediatr Transplant 2001;5:112-8.

92. Chee YL, Crawford JC, Watson HG, Greaves M. Guidelines on theassessment of bleeding risk prior to surgery or invasive procedures.British Committee for Standards in Haematology. Br J Haema-tol 2008;140:496-504.

93. Hirsh J. Reversal of the anticoagulant effects of warfarin by vitaminK1. Chest 1998;114:1505-8.

94. Hanley JP. Warfarin reversal. J Clin Pathol 2004;57:1132-9.95. Leissinger CA, Blatt PM, Hoots WK, Ewenstein B. Role of pro-

thrombin complex concentrates in reversing warfarin anticoagula-tion: a review of the literature. Am J Hematol 2008;83:137-43.

96. Schulman S, Bijsterveld NR. Anticoagulants and their reversal.Transfus Med Rev 2007;21:37-48.

97. Warkentin TE, Greinacher A, Koster A, Lincoff AM. Treatment andprevention of heparin-induced thrombocytopenia: American Collegeof Chest Physicians Evidence-Based Clinical Practice Guidelines. 8th

ed. Chest 2008;133(6 suppl):340-80S.

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

950 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

98. Selleng S, Haneya A, Hirt S, et al. Management of anticoagulation inpatients with subacute heparin-induced thrombocytopenia scheduledfor heart transplantation. Blood 2008;112:4024-7.

99. Pamboukian SV, Ignaszewski AP, Ross HJ. Management strategiesfor heparin-induced thrombocytopenia in heart-transplant candidates:case report and review of the literature. J Heart Lung Trans-plant 2000;19:810-4.

00. Laupacis A, Fergusson D. Drugs to minimize perioperative bloodloss in cardiac surgery: meta-analyses using perioperative bloodtransfusion as the outcome. The International Study of PerioperativeTransfusion (ISPOT) Investigators. Anesth Analg 1997;85:1258-67.

01. Karkouti K, Beattie WS, Dattilo KM, et al. A propensity score case-control comparison of aprotinin and tranexamic acid in high-transfu-sion-risk cardiac surgery. Transfusion (Paris) 2006;46:327-38.

02. Mahdy AM, Webster NR. Perioperative systemic haemostatic agents.Br J Anaesth 2004;93:842-58.

03. Reid RW, Zimmerman AA, Laussen PC, et al. The efficacy of tran-examic acid versus placebo in decreasing blood loss in pediatricpatients undergoing repeat cardiac surgery. Anesth Analg 1997;84:990-6.

04. Levi M, Cromheecke ME, de JE, et al. Pharmacological strategies todecrease excessive blood loss in cardiac surgery: a meta-analysis ofclinically relevant endpoints. Lancet 1999;354:1940-7.

05. Cattaneo M, Harris AS, Stromberg U, Mannucci PM. The effect ofdesmopressin on reducing blood loss in cardiac surgery—a meta-analysis of double-blind, placebo-controlled trials. Thromb Hae-most 1995;74:1064-70.

06. Steinman TI, Becker BN, Frost AE, et al. Guidelines for the referraland management of patients eligible for solid organ transplantation.Transplantation 2001;71:1189-204.

07. Dobbels F, Vanhaecke J, Desmyttere A, et al. Prevalence and corre-lates of self-reported pretransplant nonadherence with medication inheart, liver, and lung transplant candidates. Transplantation 2005;79:1588-95.

08. Martin JE, Zavala EY. The expanding role of the transplant pharma-cist in the multidisciplinary practice of transplantation. Clin Trans-plant 2004;18(suppl 12):50-4.

09. Canter C, Naftel D, Caldwell R, et al. Survival and risk factors fordeath after cardiac transplantation in infants. A multi-institutionalstudy. The Pediatric Heart Transplant Study. Circulation 1997;96:227-31.

10. Frazier EA, Naftel D, Canter C, et al. Death after cardiac transplan-tation in children: who dies, when, and why. J Heart Lung Trans-plant 1999;18:69-70.

11. Kirshbom PM, Bridges ND, Myung RJ, et al. Use of extracorporealmembrane oxygenation in pediatric thoracic organ transplantation.J Thorac Cardiovasc Surg 2002;123:130-6.

12. Fenton KN, Webber SA, Danford DA, et al. Long-term survival afterpediatric cardiac transplantation and postoperative ECMO support.Ann Thorac Surg 2003;76:843-6.

13. Mitchell MB, Campbell DN, Bielefeld MR, Doremus T. Utility ofextracorporeal membrane oxygenation for early graft failure follow-ing heart transplantation in infancy. J Heart Lung Transplant 2000;19:834-9.

14. Huddleston CB, Thul JM. Postoperative management: early graftfailure, pulmonary hypertension, and initial immunosuppressionstrategies (Chapter 7). In: Canter CE, Kirklin JK, editors. ISHLTmonograph series volume 2: pediatric heart transplantation. Addison,TX: ISHLT; 2007.

15. Duncan BW. Pediatric mechanical circulatory support. ASAIOJ 2005;51:ix-xiv.

16. Seib PM, Faulkner SC, Erickson CC, et al. Blade and balloon atrialseptostomy for left heart decompression in patients with severe ven-tricular dysfunction on extracorporeal membrane oxygenation. Cath-eter Cardiovasc Interv 1999;46:179-86.

17. Fiser SM, Tribble CG, Kaza AK, et al. When to discontinue extra-corporeal membrane oxygenation for postcardiotomy support. AnnThorac Surg 2001;71:210-4.

18. Cooper LT, Baughman KL, Feldman AM, et al. The role of endo-

myocardial biopsy in the management of cardiovascular disease: a

scientific statement from the American Heart Association, theAmerican College of Cardiology, and the European Society of Car-diology. Endorsed by the Heart Failure Society of America and theHeart Failure Association of the European Society of Cardiology.J Am Coll Cardiol 2007;50:1914-31.

19. Stehlik J, Starling RC, Movsesian MA, et al. Utility of long-termsurveillance endomyocardial biopsy: a multi-institutional analysis.J Heart Lung Transplant 2006;25:1402-9.

20. Rosenthal DN, Chin C, Nishimura K, et al. Identifying cardiac trans-plant rejection in children: diagnostic utility of echocardiography,right heart catheterization and endomyocardial biopsy data. J HeartLung Transplant 2004;23:323-9.

21. Kemkes BM, Schutz A, Engelhardt M, Brandl U, Breuer M. Nonin-vasive methods of rejection diagnosis after heart transplantation.J Heart Lung Transplant 1992;11:S221-31.

22. Horenstein MS, Idriss SF, Hamilton RM, et al. Efficacy of signal-averaged electrocardiography in the young orthotopic heart transplantpatient to detect allograft rejection. Pediatr Cardiol 2006;27:589-93.

23. Schweiger M, Wasler A, Prenner G, et al. The prognostic validity ofthe ventricular evoked response (VER) signals in cardiac transplan-tation. J Heart Lung Transplant 2005;24:1730-5.

24. Butler CR, Thompson R, Haykowsky M, Toma M, Paterson I. Car-diovascular magnetic resonance in the diagnosis of acute heart trans-plant rejection: a review. J Cardiovasc Magn Reson 2009;11:7.

25. Marie PY, Angioi M, Carteaux JP, et al. Detection and prediction ofacute heart transplant rejection with the myocardial T2 determinationprovided by a black-blood magnetic resonance imaging sequence.J Am Coll Cardiol 2001;37:825-31.

26. Geiger M, Harake D, Halnon N, Alejos JC, Levi DS. Screening forrejection in symptomatic pediatric heart transplant recipients: thesensitivity of BNP. Pediatr Transplant 2008;12:563-9.

27. Dengler TJ, Zimmermann R, Braun K, et al. Elevated serum concen-trations of cardiac troponin T in acute allograft rejection after humanheart transplantation. J Am Coll Cardiol 1998;32:405-12.

28. Arora S, Gullestad L, Wergeland R, et al. Probrain natriuretic peptideand C-reactive protein as markers of acute rejection, allograft vascu-lopathy, and mortality in heart transplantation. Transplantation 2007;83:1308-15.

29. Dengler TJ, Gleissner CA, Klingenberg R, et al. Biomarkers afterheart transplantation: nongenomic. Heart Fail Clin 2007;3:69-81.

30. Azzawi M, Hasleton PS, Turner DM, et al. Tumor necrosis factor-alpha gene polymorphism and death due to acute cellular rejection ina subgroup of heart transplant recipients. Hum Immunol 2001;62:140-2.

31. Keslar K, Rodriguez ER, Tan CD, Starling RC, Heeger PS. Comple-ment gene expression in human cardiac allograft biopsies as a cor-relate of histologic grade of injury. Transplantation 2008;86:1319-21.

32. Deng MC, Eisen HJ, Mehra MR, et al. Noninvasive discrimination ofrejection in cardiac allograft recipients using gene expression profil-ing. Am J Transplant 2006;6:150-60.

33. Pham MX, Teuteberg JJ, Kfoury AG, et al. Gene-expression profilingfor rejection surveillance after cardiac transplantation. N EnglJ Med 2010;362:1890-900

34. Rossano JW, Denfield SW, Kim JJ, et al. B-type natriuretic peptide isa sensitive screening test for acute rejection in pediatric heart trans-plant patients. J Heart Lung Transplant 2008;27:649-54.

35. Bennhagen RG, Sornmo L, Pahlm O, Pesonen E. Serial signal-aver-aged electrocardiography in children after cardiac transplantation.Pediatr Transplant 2005;9:773-9.

36. Lindenfeld J, Miller GG, Shakar SF, et al. Drug therapy in the hearttransplant recipient: part II: immunosuppressive drugs. Circula-tion 2004;110:3858-65.

37. Cantarovich M, Ross H, Arizon JM, et al. Benefit of Neoral C2 mon-itoring in de novo cardiac transplant recipients receiving basiliximabinduction. Transplantation 2008;85:992-9.

38. Barnard JB, Thekkudan J, Richardson S, et al. Cyclosporine profilingwith C2 and C0 monitoring improves outcomes after heart transplan-tation. J Heart Lung Transplant 2006;25:564-8.

39. Langers P, Press RR, den HJ, et al. Flexible limited sampling model

for monitoring tacrolimus in stable patients having undergone liver

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

951Costanzo et al. Guidelines for Heart Transplant Care

transplantation with samples 4 to 6 hours after dosing is superior totrough concentration. Ther Drug Monit 2008;30:456-61.

40. Tsunoda SM, Aweeka FT. Drug concentration monitoring of immu-nosuppressive agents: focus on tacrolimus, mycophenolate mofetiland sirolimus. BioDrugs 2000;14:355-69.

41. Knight SR, Morris PJ. Does the evidence support the use of myco-phenolate mofetil therapeutic drug monitoring in clinical practice? Asystematic review. Transplantation 2008;85:1675-85.

42. Stenton SB, Partovi N, Ensom MH. Sirolimus: the evidence for clin-ical pharmacokinetic monitoring. Clin Pharmacokinet 2005;44:769-86.

43. Brandhorst G, Tenderich G, Zittermann A, et al. Everolimus expo-sure in cardiac transplant recipients is influenced by concomitantcalcineurin inhibitor. Ther Drug Monit 2008;30:113-6.

44. Krasinskas AM, Kreisel D, Acker MA, et al. CD3 monitoring ofantithymocyte globulin therapy in thoracic organ transplantation.Transplantation 2002;73:1339-41.

45. Uber PA, Mehra MR, Park MH, Scott RL. Clopidogrel and rhabdo-myolysis after heart transplantation. J Heart Lung Transplant 2003;22:107-8.

46. Hmiel SP, Canter C, Shepherd R, Lassa-Claxton S, Nadler M. Limi-tations of cyclosporine C2 monitoring in pediatric heart transplantrecipients. Pediatr Transplant 2007;11:524-9.

47. Schubert S, bdul-Khaliq H, Lehmkuhl HB, et al. Advantages of C2monitoring to avoid acute rejection in pediatric heart transplant re-cipients. J Heart Lung Transplant 2006;25:619-25.

48. Gajarski RJ, Crowley DC, Zamberlan MC, Lake KD. Lack of corre-lation between MMF dose and MPA level in pediatric and youngadult cardiac transplant patients: does the MPA level matter? Am JTransplant 2004;4:1495-500.

49. Dipchand AI, Pietra B, McCrindle BW, Rosebrook-Bicknell HL,Boucek MM. Mycophenolic acid levels in pediatric heart transplantrecipients receiving mycophenolate mofetil. J Heart Lung Trans-plant 2001;20:1035-43.

50. Halloran PF, Gourishankar S. Principles and overview of immuno-suppression. In: Norman DJ, Laurel NJ, editors. Primer on transplan-tation. Mt Laurel, NJ: American Society of Transplantation; 2001. p.87-98.

51. Scheinman RI, Cogswell PC, Lofquist AK, Baldwin AS Jr. Role oftranscriptional activation of I kappa B alpha in mediation of immu-nosuppression by glucocorticoids. Science 1995;270:283-6.

52. Teuteberg JJ, Shullo M, Zomak R, et al. Aggressive steroid weaningafter cardiac transplantation is possible without the additional risk ofsignificant rejection. Clin Transplant 2008;22:730-7.

53. Taylor DO, Barr ML, Radovancevic B, et al. A randomized, multi-center comparison of tacrolimus and cyclosporine immunosuppres-sive regimens in cardiac transplantation: decreased hyperlipidemiaand hypertension with tacrolimus. J Heart Lung Transplant 1999;18:336-45.

54. Grimm M, Rinaldi M, Yonan NA, et al. Superior prevention of acuterejection by tacrolimus vs. cyclosporine in heart transplant recipi-ents—a large European trial. Am J Transplant 2006;6:1387-97.

55. Mancini D, Pinney S, Burkhoff D, et al. Use of rapamycin slowsprogression of cardiac transplantation vasculopathy. Circula-tion 2003;108:48-53.

56. Keogh A, Richardson M, Ruygrok P, et al. Sirolimus in de novo hearttransplant recipients reduces acute rejection and prevents coronaryartery disease at 2 years: a randomized clinical trial. Circula-tion 2004;110:2694-700.

57. Eisen HJ, Tuzcu EM, Dorent R, et al. Everolimus for the preventionof allograft rejection and vasculopathy in cardiac-transplant recipi-ents. N Engl J Med 2003;349:847-58.

58. Kobashigawa JA, Miller LW, Russell SD, et al. Tacrolimus with my-cophenolate mofetil (MMF) or sirolimus vs. cyclosporine with MMFin cardiac transplant patients: 1-year report. Am J Transplant 2006;6:1377-86.

59. Baran DA, Zucker MJ, Arroyo LH, et al. Randomized trial of tacroli-mus monotherapy: tacrolimus in combination, tacrolimus alone com-

pared (the TICTAC trial). J Heart Lung Transplant 2007;26:992-7.

60. Lehmkuhl H, Livi U, Arizon J, et al. Results of a 12-month, multi-center, randomized trial of everolimus with reduced-exposure cyclo-sporine versus mycophenolate mofetil and standard-exposure cyclo-sporine in de novo cardiac transplantation recipients. J Heart LungTransplant 2008;27(2S):S65.

61. Raichlin E, Khalpey Z, Kremers W, et al. Replacement of cal-cineurin-inhibitors with sirolimus as primary immunosuppression instable cardiac transplant recipients. Transplantation 2007;84:467-74.

62. Groetzner J, Meiser B, Landwehr P, et al. Mycophenolate mofetil andsirolimus as calcineurin inhibitor-free immunosuppression for latecardiac-transplant recipients with chronic renal failure. Transplanta-tion 2004;77:568-74.

63. Kobashigawa J, Miller L, Renlund D, et al. A randomized active-controlled trial of mycophenolate mofetil in heart transplant recipi-ents. Mycophenolate Mofetil Investigators. Transplantation 1998;66:507-15.

64. Kobashigawa JA, Tobis JM, Mentzer RM, et al. Mycophenolatemofetil reduces intimal thickness by intravascular ultrasound afterheart transplant: reanalysis of the multicenter trial. Am J Trans-plant 2006;6:993-7.

65. Kirk AD. Induction immunosuppression. Transplantation 2006;82:593-602.

66. Bonnefoy-Berard N, Revillard JP. Mechanisms of immunosuppres-sion induced by antithymocyte globulins and OKT3. J Heart LungTransplant 1996;15:435-42.

67. Bourdage JS, Hamlin DM. Comparative polyclonal antithymocyteglobulin and antilymphocyte/antilymphoblast globulin anti-CD anti-gen analysis by flow cytometry. Transplantation 1995;59:1194-200.

68. Rebellato LM, Gross U, Verbanac KM, Thomas JM. A comprehen-sive definition of the major antibody specificities in polyclonal rabbitantithymocyte globulin. Transplantation 1994;57:685-94.

69. Crespo-Leiro MG, onso-Pulpon L, Arizon JM, et al. Influence of in-duction therapy, immunosuppressive regimen and anti-viral prophy-laxis on development of lymphomas after heart transplantation: datafrom the Spanish Post-Heart Transplant Tumour Registry. J HeartLung Transplant 2007;26:1105-9.

70. Barr ML, Sanchez JA, Seche LA, et al. Anti-CD3 monoclonal anti-body induction therapy. Immunological equivalency with triple-drugtherapy in heart transplantation. Circulation 1990;82(5 suppl):IV291-4.

71. Swinnen LJ, Costanzo-Nordin MR, Fisher SG, et al. Increased inci-dence of lymphoproliferative disorder after immunosuppression withthe monoclonal antibody OKT3 in cardiac-transplant recipients.N Engl J Med 1990;323:1723-8.

72. Teuteberg J, Shullo M, Zomak R, et al. Alemtuzumab induction fa-cilitates corticosteroid-free maintenance immunosuppression inhu-man cardiac transplantation. J Heart Lung Transplant 2008;27:S201-2.

73. Pham SM, Jimenez J, Bednar BM, et al. Campath-1H in clinical hearttransplantation. J Heart Lung Transplant 2006;25:S228.

74. Das B, Shoemaker L, Recto M, Austin E, Dowling R. Alemtuzumab(Campath-1H) induction in a pediatric heart transplant: successfuloutcome and rationale for its use. J Heart Lung Transplant 2008;27:242-4.

75. Zuckermann AO, Grimm M, Czerny M, et al. Improved long-termresults with thymoglobuline induction therapy after cardiac trans-plantation: a comparison of two different rabbit-antithymocyte globu-lines. Transplantation 2000;69:1890-8.

76. Macdonald PS, Mundy J, Keogh AM, Chang VP, Spratt PM. A pro-spective randomized study of prophylactic OKT3 versus equine an-tithymocyte globulin after heart transplantation—increased morbiditywith OKT3. Transplantation 1993;55:110-6.

77. Goland S, Czer LS, Coleman B, et al. Induction therapy with thymo-globulin after heart transplantation: impact of therapy duration onlymphocyte depletion and recovery, rejection, and cytomegalovirusinfection rates. J Heart Lung Transplant 2008;27:1115-21.

78. Delgado DH, Miriuka SG, Cusimano RJ, et al. Use of basiliximaband cyclosporine in heart transplant patients with pre-operative renal

dysfunction. J Heart Lung Transplant 2005;24:166-9.

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

1

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

952 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

79. Cantarovich M, Giannetti N, Barkun J, Cecere R. Antithymocyteglobulin induction allows a prolonged delay in the initiation ofcyclosporine in heart transplant patients with postoperative renaldysfunction. Transplantation 2004;78:779-81.

80. Beniaminovitz A, Itescu S, Lietz K, et al. Prevention of rejection incardiac transplantation by blockade of the interleukin-2 receptor witha monoclonal antibody. N Engl J Med 2000;342:613-9.

81. Hershberger RE, Starling RC, Eisen HJ, et al. Daclizumab to preventrejection after cardiac transplantation. N Engl J Med 2005;352:2705-13.

82. Mehra MR, Zucker MJ, Wagoner L, et al. A multicenter, prospective,randomized, double-blind trial of basiliximab in heart transplantation.J Heart Lung Transplant 2005;24:1297-304.

83. Chin C, Pittson S, Luikart H, et al. Induction therapy for pediatric andadult heart transplantation: comparison between OKT3 and dacli-zumab. Transplantation 2005;80:477-81.

84. Segovia J, Rodriguez-Lambert JL, Crespo-Leiro MG, et al. A ran-domized multicenter comparison of basiliximab and muromonab(OKT3) in heart transplantation: SIMCOR study. Transplanta-tion 2006;81:1542-8.

85. Carlsen J, Johansen M, Boesgaard S, et al. Induction therapy aftercardiac transplantation: a comparison of anti-thymocyte globulin anddaclizumab in the prevention of acute rejection. J Heart Lung Trans-plant 2005;24:296-302.

86. Carrier M, Leblanc MH, Perrault LP, et al. Basiliximab and rabbitanti-thymocyte globulin for prophylaxis of acute rejection after hearttransplantation: a non-inferiority trial. J Heart Lung Transplant 2007;26:258-63.

87. Mattei MF, Redonnet M, Gandjbakhch I, et al. Lower risk of infec-tious deaths in cardiac transplant patients receiving basiliximab ver-sus anti-thymocyte globulin as induction therapy. J Heart LungTransplant 2007;26:693-9.

88. Meiser B, Reichart B, Adamidis I, Uberfuhr P, Kaczmarek I. Firstexperience with de novo calcineurin-inhibitor-free immunosuppres-sion following cardiac transplantation. Am J Transplant 2005;5:827-31.

89. Gonzalez-Vilchez F, de Prada JA, Exposito V, et al. Avoidance ofcalcineurin inhibitors with use of proliferation signal inhibitors in denovo heart transplantation with renal failure. J Heart Lung Trans-plant 2008;27:1135-41.

90. Lebeck LK, Chang L, Lopez-McCormack C, Chinnock R,Boucek M. Polyclonal antithymocyte serum: immune prophylaxisand rejection therapy in pediatric heart transplantation patients.J Heart Lung Transplant 1993;12:S286-92.

91. Parisi F, Danesi H, Squitieri C, Di Chiara L, Ravà L, Di Donato RM.Thymoglobuline use in pediatric heart transplantation. J Heart LungTransplant 2003;22:591-3.

92. Pollock-Barziv SM, lain-Rooney T, Manlhiot C, et al. Continuousinfusion of thymoglobulin for induction therapy in pediatric hearttransplant recipients; experience and outcomes with a novel strategyfor administration. Pediatr Transplant 2009;13:585-9.

93. Boucek RJ Jr, Naftel D, Boucek MM, et al. Induction immunother-apy in pediatric heart transplant recipients: a multicenter study.J Heart Lung Transplant 1999;18:460-9.

94. Kobashigawa JA, Katznelson S, Laks H, et al. Effect of pravastatinon outcomes after cardiac transplantation. N Engl J Med 1995;333:621-7.

95. Wenke K, Meiser B, Thiery J, et al. Simvastatin reduces graft vesseldisease and mortality after heart transplantation: a four-year random-ized trial. Circulation 1997;96:1398-402.

96. Weitz-Schmidt G, Welzenbach K, Brinkmann V, et al. Statins selec-tively inhibit leukocyte function antigen-1 by binding to a novelregulatory integrin site. Nat Med 2001;7:687-92.

97. Weis M, Pehlivanli S, Meiser BM, von SW. Simvastatin treatment isassociated with improvement in coronary endothelial function anddecreased cytokine activation in patients after heart transplantation.J Am Coll Cardiol 2001;38:814-8.

98. Kobashigawa JA, Moriguchi JD, Laks H, et al. Ten-year follow-up ofa randomized trial of pravastatin in heart transplant patients. J Heart

Lung Transplant 2005;24:1736-40.

99. Wenke K, Meiser B, Thiery J, et al. Simvastatin initiated early afterheart transplantation: 8-year prospective experience. Circula-tion 2003;107:93-7.

00. Mehra MR, Uber PA, Vivekananthan K, et al. Comparative benefi-cial effects of simvastatin and pravastatin on cardiac allograft rejec-tion and survival. J Am Coll Cardiol 2002;40:1609-14.

01. Magnani G, Carinci V, Magelli C, et al. Role of statins in the man-agement of dyslipidemia after cardiac transplant: randomized con-trolled trial comparing the efficacy and the safety of atorvastatin withpravastatin. J Heart Lung Transplant 2000;19:710-5.

02. Samman A, Imai C, Straatman L, et al. Safety and efficacy of rosu-vastatin therapy for the prevention of hyperlipidemia in adult cardiactransplant recipients. J Heart Lung Transplant 2005;24:1008-13.

03. Penson MG, Fricker FJ, Thompson JR, et al. Safety and efficacy ofpravastatin therapy for the prevention of hyperlipidemia in pediatricand adolescent cardiac transplant recipients. J Heart Lung Trans-plant 2001;20:611-8.

04. Chin C, Gamberg P, Miller J, Luikart H, Bernstein D. Efficacy andsafety of atorvastatin after pediatric heart transplantation. J HeartLung Transplant 2002;21:1213-7.

05. Hedman M, Neuvonen PJ, Neuvonen M, Holmberg C, Anti-kainen M. Pharmacokinetics and pharmacodynamics of pravastatin inpediatric and adolescent cardiac transplant recipients on a regimen oftriple immunosuppression. Clin Pharmacol Ther 2004;75:101-9.

06. Mahle WT, Vincent RN, Berg AM, Kanter KR. Pravastatin therapyis associated with reduction in coronary allograft vasculopathy inpediatric heart transplantation. J Heart Lung Transplant 2005;24:63-6.

07. Chin C, Lukito SS, Shek J, Bernstein D, Perry SB. Prevention of pe-diatric graft coronary artery disease: atorvastatin. Pediatr Trans-plant 2008;12:442-6.

08. Zuppan CW, Wells LM, Kerstetter JC, et al. Cause of death in pedi-atric and infant heart transplant recipients: review of a 20-year,single-institution cohort. J Heart Lung Transplant 2009;28:579-84.

09. Reichart B, Meiser B, Vigano M, et al. European multicenter tacroli-mus heart pilot study: three year follow-up. J Heart Lung Trans-plant 2001;20:249-50.

10. Bilchick KC, Henrikson CA, Skojec D, Kasper EK, Blumenthal RS.Treatment of hyperlipidemia in cardiac transplant recipients. AmHeart J 2004;148:200-10.

11. Heublein B, Wahlers T, Haverich A. Pulsed steroids for treatment ofcardiac rejection after transplantation. What dosage is necessary? Cir-culation 1989;80:III97-9.

12. Lonquist JL, Radovancevic B, Vega JD, et al. Reevaluation of steroidtapering after steroid pulse therapy for heart rejection. J Heart LungTransplant 1992;11:913-9.

13. Haverty TP, Sanders M, Sheahan M. OKT3 treatment of cardiac al-lograft rejection. J Heart Lung Transplant 1993;12:591-8.

14. Woodside KJ, Lick SD. Alemtuzumab (Campath 1H) as successfulsalvage therapy for recurrent steroid-resistant heart transplant rejec-tion. J Heart Lung Transplant 2007;26:750-2.

15. Onsager DR, Canver CC, Jahania MS, et al. Efficacy of tacrolimus inthe treatment of refractory rejection in heart and lung transplantrecipients. J Heart Lung Transplant 1999;18:448-55.

16. Chan MC, Kwok BW, Shiba N, et al. Conversion of cyclosporine totacrolimus for refractory or persistent myocardial rejection. Trans-plant Proc 2002;34:1850-2.

17. Yamani MH, Starling RC, Pelegrin D, et al. Efficacy of tacrolimus inpatients with steroid-resistant cardiac allograft cellular rejection.J Heart Lung Transplant 2000;19:337-42.

18. Costanzo-Nordin MR, McManus BM, Wilson JE, et al. Efficacy ofphotopheresis in the rescue therapy of acute cellular rejection inhuman heart allografts: a preliminary clinical and immunopathologicreport. Transplant Proc 1993;25:881-3.

19. Keogh AM, Arnold RH, Macdonald PS, et al. A randomized trial oftacrolimus (FK506) versus total lymphoid irradiation for the controlof repetitive rejection after cardiac transplantation. J Heart Lung

Transplant 2001;20:1331-4.

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

953Costanzo et al. Guidelines for Heart Transplant Care

20. Atluri P, Hiesinger W, Gorman RC, et al. Cardiac retransplantation isan efficacious therapy for primary cardiac allograft failure. J Cardio-thorac Surg 2008;3:26.

21. Grauhan O, Knosalla C, Ewert R, et al. Plasmapheresis and cyclo-phosphamide in the treatment of humoral rejection after heart trans-plantation. J Heart Lung Transplant 2001;20:316-21.

22. Kaczmarek I, Deutsch MA, Sadoni S, et al. Successful managementof antibody-mediated cardiac allograft rejection with combined im-munoadsorption and anti-CD20 monoclonal antibody treatment: casereport and literature review. J Heart Lung Transplant 2007;26:511-5.

23. Garrett HE Jr, Duvall-Seaman D, Helsley B, Groshart K. Treatmentof vascular rejection with rituximab in cardiac transplantation.J Heart Lung Transplant 2005;24:1337-42.

24. Rose ML, Smith J, Dureau G, Keogh A, Kobashigowa J. Mycophe-nolate mofetil decreases antibody production after cardiac transplan-tation. J Heart Lung Transplant 2002;21:282-5.

25. Itescu S, Burke E, Lietz K, et al. Intravenous pulse administration ofcyclophosphamide is an effective and safe treatment for sensitizedcardiac allograft recipients. Circulation 2002;105:1214-9.

26. Leech SH, Lopez-Cepero M, LeFor WM, et al. Management of thesensitized cardiac recipient: the use of plasmapheresis and intrave-nous immunoglobulin. Clin Transplant 2006;20:476-84.

27. Kfoury AG, Hammond ME, Snow GL, et al. Early screening forantibody-mediated rejection in heart transplant recipients. J HeartLung Transplant 2007;26:1264-9.

28. Michaels PJ, Espejo ML, Kobashigawa J, et al. Humoral rejection incardiac transplantation: risk factors, hemodynamic consequences andrelationship to transplant coronary artery disease. J Heart LungTransplant 2003;22:58-69.

29. Miller LW, Wesp A, Jennison SH, et al. Vascular rejection in hearttransplant recipients. J Heart Lung Transplant 1993;12:S147-52.

30. Brunner-La Rocca HP, Schneider J, Kunzli A, Turina M, Kio-wski W. Cardiac allograft rejection late after transplantation is a riskfactor for graft coronary artery disease. Transplantation 1998;65:538-43.

31. Klingenberg R, Koch A, Schnabel PA, et al. Allograft rejection ofISHLT grade �/�3A occurring late after heart transplantation—adistinct entity? J Heart Lung Transplant 2003;22:1005-13.

32. Esmore DS, Spratt PM, Keogh AM, Chang VP. Cyclosporine andazathioprine immunosuppression without maintenance steroids: aprospective randomized trial. J Heart Transplant 1989;8:194-9.

33. Miller LW, Wolford T, McBride LR, Peigh P, Pennington DG. Suc-cessful withdrawal of corticosteroids in heart transplantation. J HeartLung Transplant 1992;11:431-4.

34. Yamani MH, Taylor DO, Czerr J, et al. Thymoglobulin induction andsteroid avoidance in cardiac transplantation: results of a prospective,randomized, controlled study. Clin Transplant 2008;22:76-81.

35. Angermann CE, Stork S, Costard-Jackle A, et al. Reduction of cy-closporine after introduction of mycophenolate mofetil improveschronic renal dysfunction in heart transplant recipients—the IM-PROVED multi-centre study. Eur Heart J 2004;25:1626-34.

36. Rice JE, Shipp AT, Carlin JB, Vidmar SI, Weintraub RG. Late re-duction in cyclosporine dosage does not improve renal function inpediatric heart transplant recipients. J Heart Lung Transplant 2002;21:1109-12.

37. Trosch F, Rothenburger M, Schneider M, et al. First experience withrapamycin-based immunosuppression to improve kidney functionafter heart transplantation. Thorac Cardiovasc Surg 2004;52:163-8.

38. Hamour IM, Lyster HS, Burke MM, Rose ML, Banner NR. Myco-phenolate mofetil may allow cyclosporine and steroid sparing in denovo heart transplant patients. Transplantation 2007;83:570-6.

39. Gustafsson F, Ross HJ, Delgado MS, Bernabeo G, Delgado DH.Sirolimus-based immunosuppression after cardiac transplantation:predictors of recovery from calcineurin inhibitor-induced renal dys-function. J Heart Lung Transplant 2007;26:998-1003.

40. Raichlin E, Bae JH, Khalpey Z, et al. Conversion to sirolimus asprimary immunosuppression attenuates the progression of allo-graft vasculopathy after cardiac transplantation. Circulation 2007;

116:2726-33.

41. Leonard H, Hornung T, Parry G, Dark JH. Pediatric cardiac trans-plant: results using a steroid-free maintenance regimen. PediatrTransplant 2003;7:59-63.

42. Schachter AD, Meyers KE, Spaneas LD, et al. Short sirolimus half-life in pediatric renal transplant recipients on a calcineurin inhibitor-free protocol. Pediatr Transplant 2004;8:171-7.

43. van de Beek D, Kremers W, Daly RC, et al. Effect of neurologiccomplications on outcome after heart transplant. Arch Neurol 2008;65:226-31.

44. Zierer A, Melby SJ, Voeller RK, et al. Significance of neurologiccomplications in the modern era of cardiac transplantation. AnnThorac Surg 2007;83:1684-90.

45. Mayer TO, Biller J, O’Donnell J, Meschia JF, Sokol DK. Contrastingthe neurologic complications of cardiac transplantation in adults andchildren. J Child Neurol 2002;17:195-9.

46. Mateen FJ, van de BD, Kremers WK, et al. Neuromuscular diseasesafter cardiac transplantation. J Heart Lung Transplant 2009;28:226-30.

47. Kapadia SR, Nissen SE, Ziada KM, et al. Development of transplan-tation vasculopathy and progression of donor-transmitted atheroscle-rosis: comparison by serial intravascular ultrasound imaging. Circu-lation 1998;98:2672-8.

48. St Goar FG, Pinto FJ, Alderman EL, et al. Intracoronary ultrasoundin cardiac transplant recipients. In vivo evidence of “angiographicallysilent” intimal thickening. Circulation 1992;85:979-87.

49. Kobashigawa JA, Tobis JM, Starling RC, et al. Multicenter intravas-cular ultrasound validation study among heart transplant recipients:outcomes after five years. J Am Coll Cardiol 2005;45:1532-7.

50. Nicolas RT, Kort HW, Balzer DT, et al. Surveillance for transplantcoronary artery disease in infant, child and adolescent heart transplantrecipients: an intravascular ultrasound study. J Heart Lung Trans-plant 2006;25:921-7.

51. Kuhn MA, Jutzy KR, Deming DD, et al. The medium-term findingsin coronary arteries by intravascular ultrasound in infants and chil-dren after heart transplantation. J Am Coll Cardiol 2000;36:250-4.

52. Farzaneh-Far A. Electron-beam computed tomography in the assess-ment of coronary artery disease after heart transplantation. Circula-tion 2001;103:E60.

53. Pichler P, Loewe C, Roedler S, et al. Detection of high-grade steno-ses with multislice computed tomography in heart transplant patients.J Heart Lung Transplant 2008;27:310-6.

54. Dipchand AI, Bharat W, Manlhiot C, et al. A prospective study ofdobutamine stress echocardiography for the assessment of cardiacallograft vasculopathy in pediatric heart transplant recipients. PediatrTransplant 2008;12:570-6.

55. Hacker M, Hoyer HX, Uebleis C, et al. Quantitative assessment ofcardiac allograft vasculopathy by real-time myocardial contrast echo-cardiography: a comparison with conventional echocardiographicanalyses and [Tc99m]-sestamibi SPECT. Eur J Echocardiogr 2008;9:494-500.

56. Eroglu E, D’hooge J, Sutherland GR, et al. Quantitative dobutaminestress echocardiography for the early detection of cardiac allograftvasculopathy in heart transplant recipients. Heart 2008;94:e3.

57. Mehra MR. Contemporary concepts in prevention and treatment ofcardiac allograft vasculopathy. Am J Transplant 2006;6:1248-56.

58. Botha P, Peaston R, White K, et al. Smoking after cardiac transplan-tation. Am J Transplant 2008;8:866-71.

59. McDonald K, Rector TS, Braulin EA, Kubo SH, Olivari MT. Asso-ciation of coronary artery disease in cardiac transplant recipients withcytomegalovirus infection. Am J Cardiol 1989;64:359-62.

60. Simmonds J, Fenton M, Dewar C, et al. Endothelial dysfunction andcytomegalovirus replication in pediatric heart transplantation. Circu-lation 2008;117:2657-61.

61. Hussain T, Burch M, Fenton MJ, et al. Positive pretransplantationcytomegalovirus serology is a risk factor for cardiac allograft vascu-lopathy in children. Circulation 2007;115:1798-1805.

62. Kato T, Chan MC, Gao SZ, et al. Glucose intolerance, as reflected byhemoglobin A1c level, is associated with the incidence and severityof transplant coronary artery disease. J Am Coll Cardiol 2004;43:

1034-41.

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

2

3

3

3

3

3

3

3

3

954 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

63. Schroeder JS, Gao SZ, Alderman EL, et al. A preliminary study ofdiltiazem in the prevention of coronary artery disease in heart-trans-plant recipients. N Engl J Med 1993;328:164-70.

64. Erinc K, Yamani MH, Starling RC, et al. The effect of combinedAngiotensin-converting enzyme inhibition and calcium antagonismon allograft coronary vasculopathy validated by intravascular ultra-sound. J Heart Lung Transplant 2005;24:1033-8.

65. Halle AA, III, DiSciascio G, Massin EK, et al. Coronary angioplasty,atherectomy and bypass surgery in cardiac transplant recipients. J AmColl Cardiol 1995;26:120-8.

66. Aqel RA, Wells BJ, Hage FG, et al. Re-stenosis after drug-elutingstents in cardiac allograft vasculopathy. J Heart Lung Trans-plant 2008;27:610-5.

67. Radovancevic B, McGiffin DC, Kobashigawa JA, et al. Retransplan-tation in 7,290 primary transplant patients: a 10-year multi-institu-tional study. J Heart Lung Transplant 2003;22:862-8.

68. Crespo-Leiro MG, onso-Pulpon L, Vazquez de Prada JA, et al. Ma-lignancy after heart transplantation: incidence, prognosis and riskfactors. Am J Transplant 2008;8:1031-9.

69. Webber SA, Naftel DC, Fricker FJ, et al. Lymphoproliferative disor-ders after paediatric heart transplantation: a multi-institutional study.Lancet 2006;367:233-9.

70. O’Neill JO, Edwards LB, Taylor DO. Mycophenolate mofetil andrisk of developing malignancy after orthotopic heart transplantation:analysis of the transplant registry of the International Society forHeart and Lung Transplantation. J Heart Lung Transplant 2006;25:1186-91.

71. Valantine H. Is there a role for proliferation signal/mTOR inhibitorsin the prevention and treatment of de novo malignancies after hearttransplantation? Lessons learned from renal transplantation and on-cology. J Heart Lung Transplant 2007;26:557-64.

72. Senzolo M, Ferronato C, Burra P. Neurologic complications aftersolid organ transplantation. Transpl Int 2009;22:269-78.

73. Alonso EM. Long-term renal function in pediatric liver and heartrecipients. Pediatr Transplant 2004;8:381-5.

74. Lee CK, Christensen LL, Magee JC, et al. Pre-transplant risk factorsfor chronic renal dysfunction after pediatric heart transplantation: a10-year national cohort study. J Heart Lung Transplant 2007;26:458-65.

75. Wilkinson AH, Cohen DJ. Renal failure in the recipients of nonrenalsolid organ transplants. J Am Soc Nephrol 1999;10:1136-44.

76. Ojo AO. Renal disease in recipients of nonrenal solid organ trans-plantation. Semin Nephrol 2007;27:498-507.

77. Bloom RD, Doyle AM. Kidney disease after heart and lung trans-plantation. Am J Transplant 2006;6:671-9.

78. Myers BD, Newton L. Cyclosporine-induced chronic nephropathy:an obliterative microvascular renal injury. J Am Soc Nephrol 1991;2(2 suppl 1):S45-52.

79. Kopp JB, Klotman PE. Cellular and molecular mechanisms of cyclo-sporin nephrotoxicity. J Am Soc Nephrol 1990;1:162-79.

80. Schmid H, Burg M, Kretzler M, et al. BK virus associated nephrop-athy in native kidneys of a heart allograft recipient. Am J Trans-plant 2005;5:1562-8.

81. Randhawa P, Brennan DC. BK virus infection in transplant recipi-ents: an overview and update. Am J Transplant 2006;6:2000-5.

82. Gleissner CA, Murat A, Schafer S, et al. Reduced hemoglobin afterheart transplantation is no independent risk factor for survival but isassociated closely with impaired renal function. Transplanta-tion 2004;77:710-7.

83. Marchetti P. New-onset diabetes after transplantation. J Heart LungTransplant 2004;23(5 suppl):S194-201.

84. Bloom RD, Crutchlow MF. Transplant-associated hyperglycemia.Transplant Rev (Orlando) 2008;22:39-51.

85. Hathout EH, Chinnock RE, Johnston JK, et al. Pediatric post-trans-plant diabetes: data from a large cohort of pediatric heart-transplantrecipients. Am J Transplant 2003;3:994-8.

86. Pham PT, Pham PC, Lipshutz GS, Wilkinson AH. New onset diabe-tes mellitus after solid organ transplantation. Endocrinol Metab Clin

North Am 2007;36:873-90.

87. Davidson J, Wilkinson A, Dantal J, et al. New-onset diabetes aftertransplantation: 2003 International consensus guidelines. Proceedingsof an international expert panel meeting. Barcelona, Spain, 19 Feb-ruary 2003. Transplantation 2003;75(10 suppl):SS3-24.

88. Wilkinson A, Davidson J, Dotta F, et al. Guidelines for the treatmentand management of new-onset diabetes after transplantation. ClinTransplant 2005;19:291-8.

89. Kuppahally S, Al-Khaldi A, Weisshaar D, et al. Wound healing com-plications with de novo sirolimus versus mycophenolate mofetil-based regimen in cardiac transplant recipients. Am J Transplant 2006;6:986-92.

90. Diaz B, Gonzalez VF, Almenar L, et al. Gastrointestinal complica-tions in heart transplant patients: MITOS study. TransplantProc 2007;39:2397-400.

91. Kobashigawa JA, Renlund DG, Gerosa G, et al. Similar efficacy andsafety of enteric-coated mycophenolate sodium (EC-MPS, myfortic)compared with mycophenolate mofetil (MMF) in de novo hearttransplant recipients: results of a 12-month, single-blind, randomized,parallel-group, multicenter study. J Heart Lung Transplant 2006;25:935-41.

92. Galiwango PJ, Delgado DH, Yan R, et al. Mycophenolate mofetildose reduction for gastrointestinal intolerance is associated with in-creased rates of rejection in heart transplant patients. J Heart LungTransplant 2008;27:72-7.

93. Felkel TO, Smith AL, Reichenspurner HC, et al. Survival and inci-dence of acute rejection in heart transplant recipients undergoingsuccessful withdrawal from steroid therapy. J Heart Lung Trans-plant 2002;21:530-9.

94. Saeed SA, Integlia MJ, Pleskow RG, et al. Tacrolimus-associated eo-sinophilic gastroenterocolitis in pediatric liver transplant recipients:role of potential food allergies in pathogenesis. Pediatr Trans-plant 2006;10:730-5.

95. Woo SB, Treister N. Ciclosporin-induced fibrovascular polyps vs.bacillary angiomatosis. Br J Dermatol 2008;158:652-3.

96. De Iudicibus S, Castronovo G, Gigante A, et al. Role of MDR1 genepolymorphisms in gingival overgrowth induced by cyclosporine intransplant patients. J Periodontal Res 2008;43:665-72.

97. Ozdemir O, rrey-Mensah A, Sorensen RU. Development of multiplefood allergies in children taking tacrolimus after heart and livertransplantation. Pediatr Transplant 2006;10:380-3.

98. Fuchs U, Zittermann A, Berthold HK, et al. Immunosuppressive ther-apy with everolimus can be associated with potentially life-threaten-ing lingual angioedema. Transplantation 2005;79:981-3.

99. Exposito V, de Prada JA, Gomez-Roman JJ, et al. Everolimus-relatedpulmonary toxicity in heart transplant recipients. J Heart Lung Trans-plant 2008;27:797-800.

00. Lindenfeld JA, Simon SF, Zamora MR, et al. BOOP is common incardiac transplant recipients switched from a calcineurin inhibitor tosirolimus. Am J Transplant 2005;5:1392-6.

01. Lubbe J, Sorg O, Male PJ, Saurat JH, Masouye I. Sirolimus-inducedinflammatory papules with acquired reactive perforating collageno-sis. Dermatology 2008;216:239-42.

02. Deutsch MA, Kaczmarek I, Huber S, et al. Sirolimus-associated in-fertility: case report and literature review of possible mechanisms.Am J Transplant 2007;7:2414-21.

03. Raza S, Ullah K, Ahmed P, et al. Cyclosporine induced neurotoxicityin a stem cell transplant recipient. J Pak Med Assoc 2007;57:611-3.

04. Morelli N, Mancuso M, Cafforio G, Gori S, Murri L. Reversible bra-chial diplegia in a case treated with cyclosporine. Neurology 2007;69:220.

05. Sevmis S, Karakayali H, Emiroglu R, Akkoc H, Haberal M. Tacroli-mus-related seizure in the early postoperative period after liver trans-plantation. Transplant Proc 2007;39:1211-3.

06. Kaczmarek I, Schmauss D, Sodian R, et al. Late-onset tacrolimus-associated cerebellar atrophia in a heart transplant recipient. J HeartLung Transplant 2007;26:89-92.

07. Norman K, Bonatti H, Dickson RC, randa-Michel J. Sudden hearingloss associated with tacrolimus in a liver transplant recipient. Transpl

Int 2006;19:601-3.

3

3

3

33

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

955Costanzo et al. Guidelines for Heart Transplant Care

08. Frantzeskaki F, Paramythiotou E, Papathanasiou M, et al. Posteriorreversible encephalopathy syndrome in an intensive care unit patientreceiving tacrolimus. Acta Anaesthesiol Scand 2008;52:1177.

09. De Weerdt A, Claeys KG, De JP, et al. Tacrolimus-related polyneu-ropathy: case report and review of the literature. Clin Neurol Neu-rosurg 2008;110:291-4.

10. Miyagi S, Sekiguchi S, Kawagishi N, et al. Parkinsonism during cy-closporine treatment in liver transplantation: an unusual case report.Transplant Proc 2008;40:2823-2824.

11. Up to Date Online. http://www.uptodateonline.com. 2010.12. Lipshutz GS, Pham PC, Ghobrial MR, et al. Thrombotic microangi-

opathy following pancreas after kidney transplants. Clin Trans-plant 2008;22:236-41.

13. Tricot L, Lebbe C, Pillebout E, et al. Tacrolimus-induced alopecia infemale kidney-pancreas transplant recipients. Transplantation 2005;80:1546-9.

14. Kim PT, Davis JE, Erb SR, Yoshida EM, Steinbrecher UP. Colonicmalakoplakia in a liver transplant recipient. Can J Gastroen-terol 2007;21:753-5.

15. Taniai N, Akimaru K, Ishikawa Y, et al. Hepatotoxicity caused byboth tacrolimus and cyclosporine after living donor liver transplan-tation. J Nippon Med Sch 2008;75:187-91.

16. Shah S, Budev M, Blazey H, Fairbanks K, Mehta A. Hepatic veno-occlusive disease due to tacrolimus in a single-lung transplant patient.Eur Respir J 2006;27:1066-8.

17. Blanchard SS, Gerrek M, Czinn S, et al. Food protein sensitivity withpartial villous atrophy after pediatric liver transplantation with ta-crolimus immunosuppression. Pediatr Transplant 2006;10:529-32.

18. McCalmont V, Bennett K. Progressive multifocal leukoencephalop-athy: a case study. Prog Transplant 2007;17:157-60.

19. Papali A, Giannetti N, Cantarovich M. Unilateral upper extremityedema associated with sirolimus in a heart transplant patient. Trans-plantation 2007;83:240.

20. van Onna M, Geerts A, Van VH, et al. One-sided limb lymphedemain a liver transplant recipient receiving sirolimus. Acta GastroenterolBelg 2007;70:357-9.

21. Garcia-Luque A, Cordero E, Torello J, et al. Sirolimus-associatedpneumonitis in heart transplant recipients. Ann Pharmacother 2008;42:1143-5.

22. Perez MJ, Martin RO, Garcia DM, et al. Interstitial pneumonitis as-sociated with sirolimus in liver transplantation: a case report. Trans-plant Proc 2007;39:3498-9.

23. Tracey C, Hawley C, Griffin AD, Strutton G, Lynch S. Generalized,pruritic, ulcerating maculopapular rash necessitating cessation ofsirolimus in a liver transplantation patient. Liver Transpl 2005;11:987-9.

24. Zakliczynski M, Nozynski J, Kocher A, et al. Surgical wound-heal-ing complications in heart transplant recipients treated with rapamy-cin. Wound Repair Regen 2007;15:316-21.

25. Neff GW, Ruiz P, Madariaga JR, et al. Sirolimus-associated hepato-toxicity in liver transplantation. Ann Pharmacother 2004;38:1593-6.

26. Smith AD, Bai D, Marroquin CE, et al. Gastrointestinal hemorrhagedue to complicated gastroduodenal ulcer disease in liver transplantpatients taking sirolimus. Clin Transplant 2005;19:250-4.

27. Schacherer D, Zeitoun M, Buttner R, et al. Sirolimus-induced drugfever and ciclosporin-induced leukencephalopathia with seizures inone liver transplant recipient. World J Gastroenterol 2007;13:6090-3.

28. Shipkova M, Armstrong VW, Oellerich M, Wieland E. Mycopheno-late mofetil in organ transplantation: focus on metabolism, safety andtolerability. Expert Opin Drug Metab Toxicol 2005;1:505-26.

29. Eisen HJ, Kobashigawa J, Keogh A, et al. Three-year results of arandomized, double-blind, controlled trial of mycophenolate mofetilversus azathioprine in cardiac transplant recipients. J Heart LungTransplant 2005;24:517-25.

30. Kapetanakis EI, Antonopoulos AS, Antoniou TA, et al. Effect oflong-term calcitonin administration on steroid-induced osteoporosisafter cardiac transplantation. J Heart Lung Transplant 2005;24:526-

32.

31. Yong G, Hayes H, O’Driscoll G. Strategy of aggressive steroid wean-ing and routine alendronate therapy to reduce bone loss after cardiactransplantation. Transplant Proc 2007;39:3340-3.

32. Shane E, Rivas M, McMahon DJ, et al. Bone loss and turnover aftercardiac transplantation. J Clin Endocrinol Metab 1997;82:1497-506.

33. Cremer J, Struber M, Wagenbreth I, et al. Progression of steroid-associated osteoporosis after heart transplantation. Ann ThoracSurg 1999;67:130-3.

34. Lindenfeld J, Page RL, Zolty R, et al. Drug therapy in the hearttransplant recipient: part III: common medical problems. Circula-tion 2005;111:113-7.

35. Sanchez-Lazaro IJ, Martinez-Dolz L, menar-Bonet L, et al. Predictorfactors for the development of arterial hypertension following hearttransplantation. Clin Transplant 2008;22:760-4.

36. Klinge A, Allen J, Murray A, O’Sullivan J. Increased pulse wavevelocity and blood pressure in children who have undergone cardiactransplantation. J Heart Lung Transplant 2009;28:21-5.

37. Walker AH, Locke TJ, Braidley PC, Al-Mohammed A. The impor-tance of 24 hour ambulatory blood pressure monitoring after thoracicorgan transplantation. J Heart Lung Transplant 2005;24:1770-3.

38. Roche SL, Kaufmann J, Dipchand AI, Kantor PF. Hypertension afterpediatric heart transplantation is primarily associated with immuno-suppressive regimen. J Heart Lung Transplant 2008;27:501-7.

39. Brozena SC, Johnson MR, Ventura H, et al. Effectiveness and safetyof diltiazem or lisinopril in treatment of hypertension after hearttransplantation. Results of a prospective, randomized multicentertrail. J Am Coll Cardiol 1996;27:1707-12.

40. Mathias HC, Ozalp F, Will MB, et al. A randomized, controlled trialof C0- Vs C2-guided therapeutic drug monitoring of cyclosporine instable heart transplant patients. J Heart Lung Transplant 2005;24:2137-43.

41. Canter CE, Moorhead S, Saffitz JE, Huddleston CB, Spray TL. Ste-roid withdrawal in the pediatric heart transplant recipient initiallytreated with triple immunosuppression. J Heart Lung Trans-plant 1994;13:74-9.

42. Lee AH, Mull RL, Keenan GF, et al. Osteoporosis and bone morbid-ity in cardiac transplant recipients. Am J Med 1994;96:35-41.

43. Pisani B, Mullen GM. Prevention of osteoporosis in cardiac trans-plant recipients. Curr Opin Cardiol 2002;17:160-4.

44. Guo CY, Johnson A, Locke TJ, Eastell R. Mechanisms of bone lossafter cardiac transplantation. Bone 1998;22:267-71.

45. Shane E, Addesso V, Namerow PB, et al. Alendronate versus calcit-riol for the prevention of bone loss after cardiac transplantation.N Engl J Med 2004;350:767-76.

46. Leidig-Bruckner G, Hosch S, Dodidou P, et al. Frequency and pre-dictors of osteoporotic fractures after cardiac or liver transplantation:a follow-up study. Lancet 2001;357:342-7.

47. Braith RW, Magyari PM, Fulton MN, et al. Resistance exercise train-ing and alendronate reverse glucocorticoid-induced osteoporosis inheart transplant recipients. J Heart Lung Transplant 2003;22:1082-90.

48. Braith RW, Magyari PM, Fulton MN, et al. Comparison of calcitoninversus calcitonin � resistance exercise as prophylaxis for osteopo-rosis in heart transplant recipients. Transplantation 2006;81:1191-5.

49. Van Cleemput J, Daenen W, Geusens P, et al. Prevention of bone lossin cardiac transplant recipients. A comparison of biphosphonates andvitamin D. Transplantation 1996;61:1495-9.

50. Cohen A, Addesso V, McMahon DJ, et al. Discontinuing antiresorp-tive therapy one year after cardiac transplantation: effect on bonedensity and bone turnover. Transplantation 2006;81:686-91.

51. Dodidou P, Bruckner T, Hosch S, et al. Better late than never? Ex-perience with intravenous pamidronate treatment in patients with lowbone mass or fractures following cardiac or liver transplantation.Osteoporos Int 2003;14:82-9.

52. Coscia LA, Constantinescu S, Moritz MJ, et al. Report from the Na-tional Transplantation Pregnancy Registry (NTPR): outcomes ofpregnancy after transplantation. Clin Transpl 2007;29-42.

53. Sibanda N, Briggs JD, Davison JM, Johnson RJ, Rudge CJ. Preg-nancy after organ transplantation: a report from the UK Transplant

pregnancy registry. Transplantation 2007;83:1301-7.

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

3

956 The Journal of Heart and Lung Transplantation, Vol 29, No 8, August 2010

54. Transfer of drugs and other chemicals into human milk. Pediat-rics 2001;108:776-89.

55. Estes CM, Westhoff C. Contraception for the transplant patient. Se-min Perinatol 2007;31:372-7.

56. Sucato GS, Murray PJ. Gynecologic health care for the adolescentsolid organ transplant recipient. Pediatr Transplant 2005;9:346-56.

57. McKay DB, Josephson MA, Armenti VT, et al. Reproduction andtransplantation: report on the AST Consensus Conference on Repro-ductive Issues and Transplantation. Am J Transplant 2005;5:1592-9.

58. Schofield RS, Edwards DG, Schuler BT, et al. Vascular effects ofsildenafil in hypertensive cardiac transplant recipients. Am J Hyper-tens 2003;16:874-7.

59. Marconi C, Marzorati M. Exercise after heart transplantation. EurJ Appl Physiol 2003;90:250-9.

60. Biring MS, Fournier M, Ross DJ, Lewis MI. Cellular adaptations ofskeletal muscles to cyclosporine. J Appl Physiol 1998;84:1967-75.

61. Niset G, Hermans L, Depelchin P. Exercise and heart transplantation.A review. Sports Med 1991;12:359-79.

62. Keteyian S, Shepard R, Ehrman J, et al. Cardiovascular responses ofheart transplant patients to exercise training. J Appl Physiol 1991;70:2627-31.

63. Kobashigawa JA, Leaf DA, Lee N, et al. A controlled trial of exerciserehabilitation after heart transplantation. N Engl J Med 1999;340:272-7.

64. Braith RW, Schofield RS, Hill JA, Casey DP, Pierce GL. Exercisetraining attenuates progressive decline in brachial artery reactivity inheart transplant recipients. J Heart Lung Transplant 2008;27:52-9.

65. Pierce GL, Schofield RS, Casey DP, et al. Effects of exercise trainingon forearm and calf vasodilation and proinflammatory markers inrecent heart transplant recipients: a pilot study. Eur J Cardiovasc PrevRehabil 2008;15:10-8.

66. Haykowsky M, Taylor D, Kim D, Tymchak W. Exercise training im-proves aerobic capacity and skeletal muscle function in heart trans-plant recipients. Am J Transplant 2009;9:734-9.

67. Braith RW, Welsch MA, Mills RM Jr, Keller JW, Pollock ML. Re-sistance exercise prevents glucocorticoid-induced myopathy in hearttransplant recipients. Med Sci Sports Exerc 1998;30:483-9.

68. Patel JN, Kavey RE, Pophal SG, et al. Improved exercise perfor-mance in pediatric heart transplant recipients after home exercisetraining. Pediatr Transplant 2008;12:336-40.

69. Blasco LM, Parameshwar J, Vuylsteke A. Anaesthesia for noncar-diac surgery in the heart transplant recipient. Curr Opin Anaesthe-siol 2009;22:109-13.

70. Kavanagh T, Yacoub MH, Kennedy J, Austin PC. Return to workafter heart transplantation: 12-year follow-up. J Heart Lung Trans-plant 1999;18:846-51.

71. Paris W, Woodbury A, Thompson S, et al. Social rehabilitation andreturn to work after cardiac transplantation—a multicenter survey.Transplantation 1992;53:433-8.

72. White-Williams C, Jalowiec A, Grady K. Who returns to work afterheart transplantation? J Heart Lung Transplant 2005;24:2255-61.

73. Tjang YS, Tenderich G, Hornik L, Korfer R. Cardiac retransplanta-tion in adults: an evidence-based systematic review. Thorac Cardio-vasc Surg 2008;56:323-7.

74. Shuhaiber JH, Kim JB, Hur K, et al. Comparison of survival in pri-mary and repeat heart transplantation from 1987 through 2004 in theUnited States. Ann Thorac Surg 2007;83:2135-41.

75. Chin C, Naftel D, Pahl E, et al. Cardiac re-transplantation in pediat-rics: a multi-institutional study. J Heart Lung Transplant 2006;25:1420-4.

76. American Transplant Society. When to contact the transplantcenter: AST guidelines for non-transplant physicians caring forheart and/or lung transplant recipients. http://www.a-s-t.org/

index2.cfm?Section�non_transp_phys. 2010.

77. Fusar-Poli P, Picchioni M, Martinelli V, et al. Anti-depressive thera-pies after heart transplantation. J Heart Lung Transplant 2006;25:785-93.

78. De Bleser L, Matteson M, Dobbels F, Russell C, De GS. Interven-tions to improve medication-adherence after transplantation: a sys-tematic review. Transpl Int 2009;22:780-97.

79. Dew MA, DiMartini AF, De Vito DA, et al. Rates and risk factors fornonadherence to the medical regimen after adult solid organ trans-plantation. Transplantation 2007;83:858-73.

80. Havik OE, Sivertsen B, Relbo A, et al. Depressive symptoms andall-cause mortality after heart transplantation. Transplantation 2007;84:97-103.

81. Lawrence K, Stilley CS, Olshansky E, Bender A, Webber SA. Fur-ther exploration: maturity and adherence in adolescent and youngadult heart transplant recipients. Prog Transplant 2008;18:50-4.

82. Szepietowski JC, Reich A, Nowicka D, Weglowska J, Szepietows-ki T. Sun protection in renal transplant recipients: urgent need foreducation. Dermatology 2005;211:93-7.

83. Wray J, Waters S, Radley-Smith R, Sensky T. Adherence in adoles-cents and young adults following heart or heart-lung transplantation.Pediatr Transplant 2006;10:694-700.

84. Dobbels F, Van Damme-Lombaert R, Vanhaecke J, De GS. Growingpains: non-adherence with the immunosuppressive regimen in ado-lescent transplant recipients. Pediatr Transplant 2005;9:381-90.

85. Keith DS, Cantarovich M, Paraskevas S, Tchervenkov J. Recipientage and risk of chronic allograft nephropathy in primary deceaseddonor kidney transplant. Transpl Int 2006;19:649-56.

86. Rosen DS, Blum RW, Britto M, Sawyer SM, Siegel DM. Transitionto adult health care for adolescents and young adults with chronicconditions: position paper of the Society for Adolescent Medicine. JAdolesc Health 2003;33:309-11.

87. Williams MJ, Lee MY, DiSalvo TG, et al. Biopsy-induced flail tri-cuspid leaflet and tricuspid regurgitation following orthotopic cardiactransplantation. Am J Cardiol 1996;77:1339-44.

88. Nguyen V, Cantarovich M, Cecere R, Giannetti N. Tricuspid regur-gitation after cardiac transplantation: how many biopsies are toomany? J Heart Lung Transplant 2005;24(7 suppl):S227-31.

89. Wiklund L, Suurkula MB, Kjellstrom C, Berglin E. Chordal tissue inendomyocardial biopsies. Scand J Thorac Cardiovasc Surg 1994;28:13-8.

90. Mielniczuk L, Haddad H, Davies RA, Veinot JP. Tricuspid valvechordal tissue in endomyocardial biopsy specimens of patients withsignificant tricuspid regurgitation. J Heart Lung Transplant 2005;24:1586-90.

91. Levi DS, DeConde AS, Fishbein MC, et al. The yield of surveillanceendomyocardial biopsies as a screen for cellular rejection in pediatricheart transplant patients. Pediatr Transplant 2004;8:22-8.

92. Billingham ME, Cary NR, Hammond ME, et al. A working formu-lation for the standardization of nomenclature in the diagnosis ofheart and lung rejection: Heart Rejection Study Group. The Interna-tional Society for Heart Transplantation. J Heart Transplant 1990;9:587-93.

93. Winters GL. The challenge of endomyocardial biopsy interpretationin assessing cardiac allograft rejection. Curr Opin Cardiol 1997;12:146-52.

94. Cai J, Terasaki PI. Humoral theory of transplantation: mechanism,prevention, and treatment. Hum Immunol 2005;66:334-42.

95. Delgado JF, Sanchez V, de la Calzada CS. Acute rejection after hearttransplantation. Expert Opin Pharmacother 2006;7:1139-49.

96. Bierl C, Miller B, Prak EL, et al. Antibody-mediated rejection inheart transplant recipients: potential efficacy of B-cell depletion and

antibody removal. Clin Transpl 2006;489-96.