Thomas 2014

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Chapter 28

Current Status of Heart Transplantation


Sunu S. Thomas and Mandeep R. Mehra
The Center for Advanced Heart Disease, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA

Chapter Outline
28.1 The Heart Failure Syndrome 403 28.6.6 Hyperacute Rejection 411
28.2 Cardiac Transplantation: Historical Achievement 405 28.6.7 Acute Graft Rejection 411
28.3 Heart Transplant Evaluation 406 28.7 Immunosuppression Strategies and Rejection
28.3.1 Age and Comorbidites 406 Surveillance 411
28.3.2 Chronic Kidney Disease 407 28.7.1 Induction Therapy 412
28.3.3 Malignancy 407 28.8 Maintenance Immunosuppression 413
28.3.4 Infection 407 28.8.1 Corticosteroids 413
28.3.5 Pulmonary Hypertension 407 28.8.2 Calcineurin Inhibitors 413
28.3.6 Psychosocial Functioning 408 28.8.3 Antiproliferative Agents 414
28.3.7 Sensitization 408 28.8.4 mTOR Inhibitors 414
28.4 Listing Status 408 28.9 Rejection Surveillance 414
28.5 The Ideal Donor Heart 409 28.9.1 Endomyocardial Biopsy 414
28.5.1 Donor Characteristics 409 28.9.2 Biomarker Tests 415
28.5.2 Evaluation of the Donor Heart 409 28.10 Transplantation Outcomes 415
28.6 Perioperative Management 409 28.10.1 Cardiac Allograft Vasculopathy 415
28.6.1 Graft Preservation and Optimizing Ischemic 28.11 Mechanical Horizon: VADs as a Bridge to
Time 409 Transplant 417
28.6.2 Early Postoperative Care 410 28.12 Future Directions in Cardiac Transplantation 419
28.6.3 Cardiac Allograft Function 410 28.13 Conclusion 419
28.6.4 Right Ventricular Failure 410 Acknowledgments 419
28.6.5 Primary Graft Failure 410 References 419

28.1 THE HEART FAILURE SYNDROME from poor ventricular compliance leading to impairment
There are nearly 6 million individuals in the United of diastolic parameters, termed Heart Failure with
States with a diagnosis of heart failure. The annual inci- Preserved Ejection Fraction (HFpEF). Regardless of eti-
dence approximates 600,000 new cases, which is further ology, patients presenting with signs and symptoms of
complicated by a high hospitalization rate and yearly heart failure may do so acutely with de novo ventricular
mortality rate in excess of 30% in patients with advanced dysfunction, or as an exacerbation of a known chronic
disease. With economic costs continuing to spiral out of heart failure condition. Patients universally complain of
control, the burden and severity of this epidemic cannot similar symptoms including dyspnea, fatigue, presyncope,
be understated [1]. syncope, orthopnea, and paroxysmal nocturnal dyspnea
The cardinal feature of heart failure is the elevation of reflecting the consequences of impaired circulatory flow
ventricular filling pressures that eventually lead to a com- and overall fluid retention. Physical examination typically
promised cardiac output. This may arise from poor sys- reveals hallmark findings of volume overload including
tolic function resulting from impairment of ventricular pulmonary congestion, jugular venous distension, increas-
contractility, termed Heart Failure with Reduced Ejection ing abdominal girth and peripheral edema, and signs of
Fraction (HFrEF). Alternatively, heart failure may arise poor perfusion including relative hypotension, cool

Regenerative Medicine Applications in Organ Transplantation.


© 2014 Elsevier Inc. All rights reserved. 403
404 PART | IV Heart

Advanced Heart Failure


NYHA Class IIIb / IV Symptoms

Optimize Neurohormonal Blockade Cardiac Resynchronization Therapy


Beta-Blockers, ACE inhibitors, ARBs, Aldosterone antagonists LVEF < 35%, QRS ≥ 120 ms (ideally QRS ≥ 150 ms, LBBB, NSR)

Medical Futility
Intolerance of medical therapy due to hypotension
Initiation of inotropes for systemic perfusion
Worsening end-organ function BUN ≥ 40 mg/dl, rising Cre)
Diuretic resistance
Persistent hyponatremia (< 136 mEq/L)

Refractory symptoms
>2 Hospitalizations/year

Poor predicted survival


( www.seattleheartfailuremodel.org)

Evaluation for Advanced Cardiac Therapies


Referral to heart failure specialist
Candidacy for heart replacement therapy
(Cardiac Transplant vs. Mechanical Circulatory Support vs. Palliation)

FIGURE 28.1 Algorithm for medical decision-making in patients with end-stage heart failure. Modified from Mehra et al. [23].

extremities, and end-organ dysfunction. These bedside It appears that regardless of inciting injury or disease
findings may be further categorized into four prognosti- process, all cardiomyopathies share a common pathophys-
cally important hemodynamic profiles on the basis of iological pathway. While our current therapies target the
adequacy of perfusion (warm versus cold) and measure of mediators of neurohormonal imbalance, for reasons still
volume overload (dry versus wet) [2]. Patients who are poorly understood, the impaired ventricle negatively
“cold and wet” represent the most advanced stages of dis- remodels through a process of dilatation and myocardial
ease and have a 1 year increased risk of death and need thinning [21]. Clinically, this amounts to persistence and
for cardiac transplantation which is also two times worse worsening of symptoms, deteriorating ventricular func-
than those who are “warm and dry” [3,4]. tion, hemodynamic and electrical instability, and frequent
The management of heart failure has evolved over the rehospitalizations, culminating in a medically refractory
course of the last two decades toward an evidence-based state and inevitable death [22].
strategy incorporating the use of beta-blockers, angioten- Stage D heart failure, characterized by a refractory
sin converting enzyme inhibitors, or angiotensin receptor state of structural heart disease, unrelenting symptomol-
blockers and aldosterone antagonists [5 15]. In addition, ogy and grave prognosis, often requires the escalation of
survival and quality of life have significantly improved medical therapies [1] (Figure 28.1). This typically
with the use of implantable defibrillators and cardiac requires incremental use of diuretics to relieve conges-
resynchronization therapy in eligible patients. However, tion, and for the hemodynamically compromised patient,
the clinical challenge lies in the relative ceiling effect of the judicious use of inotropic therapy either for
the benefits derived from these medical and device-based temporary relief of congestion or as a longer-term
therapies [16 18]. Although mortality rates have bridging or palliation strategy to augment a low cardiac
improved by the use of disease modifying therapy, with output state. However, symptomatic relief with such
few exceptions such as fulminant myocarditis or a peri- medications does not improve prognosis or reverse the
partum cardiomyopathy, the etiology of the underlying downward spiral leading to disease progression, and
cardiomyopathy is generally nonreversible [19,20]. may hasten patient mortality [24,25]. For some patients,
Chapter | 28 Current Status of Heart Transplantation 405

FIGURE 28.2 Cardiac transplan-


tation trends according to time and
region. Figure reprinted from [33].

marginal blood pressures necessitate the withdrawal of With the development of cardiopulmonary bypass,
medications, such as beta-blockers and antagonists of the improved organ preservation with cardioplegia, and
renin angiotensin aldosterone system, originally pre- refinement of surgical technique including orthotopic
scribed for their mortality benefit. For others too hemody- transplantation, early investigators were able to improve
namically compromised, ventricular assist devices graft survival in animal models and recognize the need
(VADs) have provided a necessary bridge to transplanta- for improved immunosuppression.
tion for mechanical circulatory support [26,27]. However, The success of the first human cardiac transplant was
the arduous path traveled by the heart failure patient followed promptly by both a surge in optimism and in the
eventually reaches a crossroad at which point a decision number of surgeries to at least 100 in 17 countries,
must be made jointly by the patient and their advanced including the first US transplantation by Dr. Norman
cardiac care providers as to whether to pursue a path of Shumway 1 month after Dr. Barnaard’s achievement.
palliation and comfort, or to engage in the process leading However, poor posttransplant survival, attributed to poor
to possible cardiac transplantation. allograft survival and opportunistic infections, resulted in
This chapter will outline our current progress and contraction in both the number of transplant surgeries and
understanding of cardiac transplantation as a heart programs offering them. Yet, not unlike many innovations
replacement therapy for selected patients with end-stage in modern medicine requiring refinement of process, the
heart failure. field was revitalized with the development of endomyo-
cardial biopsies for rejection surveillance [31], improved
immunosuppression strategies primarily involving the use
28.2 CARDIAC TRANSPLANTATION: of cyclosporine [32], and the formation of the United
Network for Organ Sharing (UNOS) to proctor the pro-
HISTORICAL ACHIEVEMENT curement and allocation of donor hearts to eligible
The first successful human cardiac transplantation by Dr. recipients.
Christian Baarnard in 1967 ushered in the modern age of According to the most recent 2012 report from the
heart replacement therapy [28]. This achievement was the Registry of the International Society for Heart and Lung
result of decades of earnest investigation examining surgi- Transplantation, nearly 4000 heart transplants are now
cal technique and the biology of organ rejection. performed worldwide annually, consistent with trends
Beginning in 1905, the first case of a successful heart over the last decade (Figure 28.2) [33]. Overall, more
transplant involved the implantation of a canine heart into than 100,000 heart transplants have been performed since
the neck of a canine recipient [29]. The transplant donor Dr. Barnard’s seminal surgery. Unfortunately, however,
organ was anastomosed heterotopically to the recipient’s there continues to be great imbalance in the available
cardiopulmonary circulation without removal of the donor heart pool and the number of patients requiring
native heart. In 1964, Dr. James Hardy performed the first them, with those in need numbering close to half a mil-
successful xenotransplantation in which the heart of a lion in the United States alone [1]. In fact, the number of
chimpanzee was transplanted into a human and was func- patients active on the transplant list has grown by 19.2%
tional for 90 minutes at which point it began to fail [30]. since 2005 [34].
406 PART | IV Heart

and years of life. The challenge for the advanced cardiac


TABLE 28.1 Listing Criteria for Cardiac Transplant care team is in the identification of those patients who
Indications will be both eligible and capable of benefitting from a
valuable, yet limited, resource.
Anticipated poor 1 year survival
Advanced symptoms (NYHA Class IV) refractory to medical Based on recently published International Society of
therapy Heart and Lung Transplant (ISHLT) registry data, the
Peak VO2 , 12 mL/kg/min with beta-blockers; ,14 mL/kg/min typical transplant patient can be characterized as being
without beta-blockers male (76.3%), approximately 54 years of age with an
Refractory angina not amenable to revascularization
underlying nonischemic cardiomyopathy (53.8%), and
Intractable arrhythmia not amenable to pharmacotherapy,
ablation, or defibrillator therapy with medical comorbidities including diabetes (25.4%),
hypertension (44.1%), and prior cardiac surgery (45.4%).
Contraindications
The relative medical acuity of the candidate patient await-
Absolute ing transplant is high, as inferred by the use of intrave-
Fixed pulmonary hypertension nous inotropes (42.6%) and nearly a majority being
Pulmonary artery systolic pressure .60 mmHg hospitalized at the time of transplantation (44.6%).
Pulmonary vascular resistance .6 Wood units Patients requiring left ventricular mechanical support
Transpulmonary gradient .15 mmHg accounted for 27.3% of transplant patients.
Concomitant illness adversely limiting life expectancy
(,2 years)
A challenge in prognosticating patients with heart fail-
Malignancy within 5 years ure is the lack of concordance between clinical symp-
Persistent renal failure despite toms, left ventricular function, and overall functional
medication adjustments or capacity. In fact, left ventricular ejection fraction is not
augmentation of perfusion with inotropes or circulatory itself a sole listing criteria for cardiac transplant. In 1991,
support
Irreversible hepatic dysfunction
Mancini and colleagues introduced the utility of peak
Severe obstructive pulmonary disease (FEV1 ,1 L/min) oxygen consumption as an objective measure of cardiac
AIDS with recurrent opportunistic infection performance and transplant eligibility [35]. In their semi-
Active disease with multisystem involvement, e.g., SLE, nal work, they found that a peak VO2 less than 14 mL/kg/
amyloidosis, sarcoidosis min was associated with a 1 year mortality rate of less
Relative than 50%. By providing a diagnostic cut-off value, they
Age .72 years were able to conclude that cardiac transplant could be
Morbid obesity (BMI . 35 kg/m2) safely postponed in patients with a peak VO2 greater than
Diabetes with end-organ involvement 14 mL/kg/min, as their 1 year survival rate was 94%. The
Significant cerebrovascular or peripheral vascular disease peak VO2 may be further modified by patient characteris-
Psychosocial issues
tics including age, gender, and race, and also by specific
Active substance abuse
Active mental illness therapies such as beta-blockers [36 39]. Accordingly,
Medical noncompliance cardiac transplantation may be indicated in patients
Current smoking habit with a peak VO2 of less than 14 mL/kg/min, or if on
Modified from Mehra et al. [41] and Leitz and Mancini [40].
concurrent beta-blocker therapy, a peak VO2 less than
12 mL/kg/min.
Ultimately, a heart failure patient’s candidacy for
transplant is not as dependent on the burden of indication
28.3 HEART TRANSPLANT EVALUATION
but on the identification of any contraindication that may
The heart failure patient being evaluated for cardiac trans- impair graft function and the recipient’s quality of life
plantation will typically have a dire prognosis. Death is following transplant [40,41]. Table 28.1 highlights the list
anticipated within 1 year despite optimization of medical of indications and contraindications that may determine
therapies that may alter symptom status, ventricular func- patient eligibility for a heart transplant. As such, each
tion, or electrical and hemodynamic instability. patient must proceed through a battery of clinical investi-
Ambulatory patients typically describe severe functional gations, including a thorough evaluation of their psycho-
limitation with NYHA Class IIIb or IV symptoms. The logical well-being and social circumstances prior to
severity of hemodynamic compromise is obvious in transplant listing.
patients with cardiogenic shock requiring hemodynamic
support using inotropes, intra-aortic balloon pumps, or
mechanical circulatory assist devices. For such patients, a
28.3.1 Age and Comorbidites
cardiac transplant may provide a solution to an otherwise Patients aged over 70 years are not typically considered
irreversible process with the hope of an improved quality for cardiac transplant, owing primarily to age-associated
Chapter | 28 Current Status of Heart Transplantation 407

medical comorbidities and anticipated limited life expec- of remission status [42]. More recent guidelines suggest
tancy. However, with longer waiting times for a trans- that at least in some situations where survival from the
plant, there is a concern regarding the distinction and cancer is expected to outlast the median survival from
appropriateness of listing age versus age at transplant that cardiac transplantation, careful consideration to bending
will need to be addressed with a growing and aging heart the 5-year remission rule may be followed in close con-
failure population. Screening for medical comorbidities sultation with an oncological team.
includes diabetes with end-organ complications, severe
pulmonary disease (FEV ,40%), liver cirrhosis, severe
peripheral vascular disease, and osteoporosis which may
28.3.4 Infection
complicate perioperative outcomes and limit overall life
expectancy. For similar reasons, caution is exercised in Patients with an active infection at the time of evaluation
listing patients with extremes of body mass index are typically deemed transplant ineligible. An evolving
(BMI . 35 kg/m2 and ,18 kg/m2). However, age alone is perspective in the field has involved the eligibility for
no longer an absolute contraindication for cardiac trans- patients who possess chronic viral infections, e.g., human
plantation with several centers having expanded their age immunodeficiency virus (HIV). The risk of opportunistic
limits or considered using alternative lists where older or infections with immunosuppression, potential drug inter-
marginal donors who may otherwise not be used for tradi- actions with antiretroviral therapy, and presumed limits to
tional candidates are paired with older recipients. HIV-related life expectancy represent a sampling of rea-
sons explaining the relatively few cases of heart trans-
plantation in patients with the disease [43]. However,
28.3.2 Chronic Kidney Disease renal transplant outcomes in HIV-positive patients are
comparable to those kidney recipients not infected with
Renal failure is common among patients with end-stage the virus [44]. Personal communications from transplant
heart failure stemming from systemic disease including centers suggest that candidacy for heart transplant may be
diabetes, hypertension, and atherosclerosis, or from limited by under-referral for advanced cardiac therapies
complex cardiorenal interactions. Efforts to ensure that stemming from prohibitive comorbidities and psychoso-
renal function is not a consequence of impaired perfu- cial circumstances.
sion typically employ a trial of inotropic therapy, an Outcomes in cardiac transplant patients with a preex-
intra-aortic balloon pump, or mechanical circulatory isting history of chronic hepatitis B or C viral infection
support. However, patients with a persistently elevated may be attributed more to the progression of liver disease
creatinine ( . 2.0 mg/dL), particularly with significant rather than allograft failure [45,46]. Routine transplant
proteinuria, are deemed ineligible for a heart transplant evaluations in such patients may involve a liver biopsy to
in isolation. A key posttransplant issue is the near auto- assess for pathological cirrhosis, or a serum measurement
matic deterioration of renal function as a consequence of viral titers. In the end, however, transplant candidacy
of particular immunosuppressive medications, particu- may be determined largely by center experience.
larly calcineurin inhibitors, and its associated risk with
long-term survival. Therefore, collaborative efforts with
renal specialists may be necessary for consideration of
renal biopsies to delineate underlying kidney pathology 28.3.5 Pulmonary Hypertension
and for potential combined listing for both heart and
Prior to listing, patients undergo right heart catheterization
kidney transplant. The most recent ISHLT registry data
for the assessment of pulmonary pressures. An absolute
suggest that heart kidney transplants have increased,
contraindication, fixed pulmonary arterial hypertension
but still remain low with a total of only 67 such cases
can lead to the development of acute right ventricular fail-
in 2010 [33].
ure of the donor heart not accustomed to the severity of
such an afterload. The degree of reversible pulmonary
hypertension is evaluated using pulmonary vasodilators,
28.3.3 Malignancy including adenosine, prostacyclin, nitroprusside, or nitric
All patients undergo age-related cancer screening includ- oxide; or inodilators such as milrinone or dobutamine.
ing colonoscopy, mammography, and prostate evaluation. Pulmonary hypertension is contraindicated in patients who
Patients with a history of malignancy require at least 5 maintain a transpulmonary gradient greater than 15, a pul-
years of remission prior to consideration for transplant. monary systolic pressure greater than 50 mmHg or the
However, the predisposition to skin and lymphoprolifera- indexed pulmonary vascular resistance exceeds 6 Woods
tive cancers by posttransplant immunosuppressive thera- units, despite a vasodilator challenge while maintaining a
pies may render a patient transplant ineligible regardless systemic pressure greater than 85 mmHg [41].
408 PART | IV Heart

TABLE 28.2 UNOS Status Definitions for Cardiac Transplant Listing According to Medical Urgency

Status Definition
Status 1A A. Acute inpatient mechanical circulatory support
Left and/or right Ventricular Assist Device ( VAD)a
Total Artificial Heart (TAH)
Intra-aortic balloon pump
Extracorporeal Membrane Oxygenation (ECMO)
B. Mechanical circulatory support with device-related complications
Thromboembolism
Device infection
Mechanical failure
Life-threatening ventricular arrhythmias
“Other” complications with approval from a respective cardiac transplant regional review board
C. Continuous mechanical ventilation
D. Continuous infusion of either
i. a single high-dose intravenous inotrope or
ii. multiple intravenous inotropes
plus continuous hemodynamic monitoring of intracardiac pressures using a pulmonary artery catheter

Status 1A Exception Medical urgency despite failure to meet any of the aforementioned criteria
Requires approval from a respective cardiac transplant regional review board
Status 1B Transplant candidate discharged home with either
i. left and/or right VAD or
ii. continuous infusion of intravenous inotropes.
Status 2 Transplant candidate who does not meet the criteria for Status 1A or 1B
Status 7 Inactive on the transplant list despite eligibility due to prohibitive medical or social circumstance
a
Eligible to maintain 1A status for up to 30 days postimplantation.

28.3.6 Psychosocial Functioning their risk for the development of preformed antibodies
against donor human leukocyte antigens (HLA). Prior to
Psychological health and social circumstances are major active transplant listing, programs often subject their sen-
determinants of transplant eligibility, even after medical sitized patients to immunomodulating therapies to lessen
clearance. Active substance abuse is an absolute contrain- the risk of anticipated rejection within a given donor
dication to transplant. While programs vary in their absti- pool. These strategies include intravenous immunoglobu-
nence periods, patients are typically challenged to remain lin, plasmapheresis, and rituximab to reduce the burden
substance free for a period of 6 months. The data is clear of preformed antibodies, and in so doing, desensitize the
in demonstrating that smoking has a deleterious impact patient.
on graft rejection and mortality posttransplant [47].
Psychological function and overall emotional stability are
evaluated due to their relationship with adherence to med-
ical regimens and self-care, which ultimately impact 28.4 LISTING STATUS
rejection status, morbidity, and mortality following trans- Once deemed eligible, patients in the United States are
plant [48]. listed for cardiac transplant with a priority status in keep-
ing with their medical urgency in accordance with UNOS
guidelines. As highlighted in Table 28.2, Status 1A
28.3.7 Sensitization patients are considered of the highest priority due to the
Rejection adversely affects transplant outcomes. As will severity of their overall clinical acuity. These ICU-bound
be outlined, rejection may be cellular or antibody- patients typically require hemodynamic support with a
mediated or mixed, with the consequence of graft dys- mechanical circulatory assist device, an intra-aortic bal-
function or more severely, overt graft failure. Antigenic loon pump, or multiple inotropes with an in situ pulmo-
exposure through blood products, pregnancy, VADs, and nary artery catheter for hemodynamic tailoring. Status 1B
prior transplantation sensitizes the recipient by increasing may require the use of a mechanical circulatory assist
Chapter | 28 Current Status of Heart Transplantation 409

device but are typically ambulatory and not admitted to 28.5.2 Evaluation of the Donor Heart
hospital. Status 2 patients do not meet criteria for Status
1A or 1B listing, and Status 7 patients are typically those For those hearts obtained from donors older than 40 years
eligible for transplant but not active on the list due to the of age, concomitant coronary artery disease should be
development of a clinical circumstance that would make ruled out due to its predilection for the development of
either the surgery or posttransplant outcome deleterious, coronary allograft vasculopathy and graft failure.
including active infection, stroke, or prohibitive obesity. Assessment is made by a coronary angiogram, or if an
invasive evaluation is not feasible, by direct palpation of
the coronary arteries during organ procurement surgery.
While valvular disease is not a contraindication to trans-
28.5 THE IDEAL DONOR HEART plant, surgical repair, or replacement of the affected valve
can be undertaken immediately prior to transplantation.
The availability of a donor organ is typically the result of
Echocardiography is an important screening tool used to
a fatal accident or self-induced trauma leading to brain
evaluate the donor heart structure and function. Typically,
death, but with preserved viability of the heart. Despite
regional wall motion abnormalities must be further evalu-
the limited number of donor organs available, appropriate
ated for related coronary artery disease or an underlying
selection is still necessary to ensure favorable posttrans-
cardiomyopathy in the donor heart. Ventricular hypertro-
plant outcomes for the recipient. At the time of organ
phy has been attributed to early graft failure and mortality
donation, the potential donor heart is evaluated for mar-
posttransplant, particularly if the ventricular thickness
kers of abnormal function and the medical comorbidities
exceeds 14 mm. Even though systolic dysfunction may be
of the deceased. While no single parameter ultimately dis-
considered a contraindication, its presence may reflect the
qualifies a potential donor, consensus must be reached by
transient catecholaminergic imbalance attributed to the
the transplant team taking into account the totality of
patient’s clinical condition and mechanism of death lead-
such variables.
ing to global myocardial ischemia and ventricular
impairment. Current recommendations prohibit the use of
a potential donor heart requiring escalating inotropic ther-
28.5.1 Donor Characteristics apy such as dopamine 20 μg/kg/min or equivalent doses
Initial considerations focus on donor age, mechanism of of other catecholamines [42].
death, high-risk social behaviors, and an ongoing assess- Another important consideration for an orthotopic car-
ment of the clinical requirements to ensure preservation diac transplantation is ensuring that the donor heart
of organ function in the deceased. Even though an upper matches the size and hemodynamic needs of the recipient.
age limit has not been defined, current guidelines recom- Significant complications arise if a donor-recipient mis-
mend the use of donor hearts less than 45 years of age, match occurs. A large donor heart may not permit closure
namely due to the concern for age-related changes to the of the chest wall, and an undersized heart may not ade-
heart that may impact long-term graft function. However quately perfuse a larger recipient. Consequently, current
due to donor organ scarcity it is not uncommon to extend guidelines recommend that the weight of donor patient be
this boundary. Death of the donor typically arises from no less than 30% below that of the recipient [42].
toxic overdoses or brain death resulting from trauma,
anoxia, or fatal cerebrovascular accident. Although some
studies suggest that traumatic brain injury may portend 28.6 PERIOPERATIVE MANAGEMENT
poorer long-term survival among transplant recipients, 28.6.1 Graft Preservation and Optimizing
such a mechanism of death is nonprohibitive [49,50].
However, there is greater concern for death resulting
Ischemic Time
from toxicity as the transplant outcome data are limited. A major determinant of early transplant outcomes and
Drug abuse, incarceration, and lifestyle practices that pre- overall right ventricular function is the duration of ische-
dispose to communicable disease such as HIV and hepati- mic time. Typically, measured from the instance of aortic
tis B and C, must be explored. Current consensus enables cross-clamping at the time of organ procurement until
the use of donor hearts from individuals involved in such which time the donor heart is perfused within the recipi-
high-risk social behaviors as the long-term outcomes ent, there is consensus that this duration not exceed 4 h.
from smaller studies are not necessarily worse. However, This single variable has helped form the basis of geo-
due to the implicit transmissible infectious risks derived graphical territories from which a given transplant candi-
from such patients, careful screening is necessary at the date’s donor pool resides.
time of transplant evaluation, in addition to informed con- The donor heart is immediately subjected to hypoxic-
sent from the recipient. ischemic injury at the time of explantation, followed by
410 PART | IV Heart

the risk of reperfusion injury in the posttransplant period. recommended to maintain heart rates greater than
Current strategies for graft preservation focus on curbing 90 bpm. Atrioventricular pacing may be required in the
the perturbations in cellular, tissue, and metabolic distress event of medical nonresponsiveness [42].
associated with these surgical events. To this end, the Persistent hypotension necessitates evaluation for car-
donor organ is maintained hypothermic by direct applica- diac tamponade, graft dysfunction, and contributes to a
tion of ice and a cold perfusate, such as the University of vasodilatory state such as infection.
Wisconsin preservation solution consisting of potassium,
magnesium, and free radical scavengers, lactobionate and
raffinose, to achieve homeostasis of electrolytes and limit
28.6.3 Cardiac Allograft Function
myocardial injury [51]. For the transplant patient, there is heightened vigilance
Investigations are ongoing with the use of perfusion for early graft dysfunction attributed to either (i) right
systems to improve donor organ integrity and preserva- ventricular failure, (ii) primary graft failure, (iii) hyper-
tion across longer ischemic times. Already approved in acute rejection, or (iv) acute cellular rejection.
Europe, the Transmedics Organ Care System is currently
being studied to compare its perfusion system that sup-
plies the donor heart with warm, nutrient-enriched, oxy-
28.6.4 Right Ventricular Failure
genated blood with that of conventional cold preservation Right ventricular dysfunction of the donor heart may arise
techniques. Such strategies may potentially reduce ische- as a manifestation of primary graft failure. However, high
mic injury and broaden the donor pool across a wider pulmonary vascular pressures, excessive volume loading,
geographical region for eligible transplant candidates. and periprocedural factors including direct injury and pro-
longed ischemic times can compromise the right ventricu-
lar function of a normal donor heart. In addition, multiple
28.6.2 Early Postoperative Care factors can lead to an acute rise in pulmonary vascular
As with all high-risk surgical patients, the early posttrans- resistance in recipients with no prior history of pulmonary
plant period can be marked by challenges in cardiopulmo- arterial hypertension. Increased pulmonary vascular tone
nary stability, surgical bleeding, infection, and recovery of can develop from hypoxemia, unrecognized intracardiac
end-organ function. Generation of an adequate cardiac shunts and postoperative lung atelectasis, pleural effu-
output and perfusion pressure may be limited by allograft sions, and infection. Defined hemodynamically, right ven-
function and postoperative vasodilatory shock. In addition tricular failure is characterized by an elevated right atrial
to preexisting comorbidities, ischemic time, and quality of pressure (.20 mmHg), and a low cardiac output in
graft preservation, historical evidence suggests that donor the absence of high left ventricular filling pressures.
heart inotropic and chronotropic reserve may be furthered Additional features include hepatic congestion marked by
impaired by sympathetic denervation and myocardial a transaminitis and hyperbilirubinemia, worsening renal
depletion of stored catecholamines [52,53]. Invasive moni- failure, and normal to low pulmonary pressures due to
toring using pulmonary catheters may facilitate hemody- poor systolic function of the right ventricle. Management
namic tailoring according to intracardiac and systemic is typically supportive while monitoring for ventricular
pressures, mixed venous oxygen saturations, and recovery. Strategies range from right-sided afterload
calculated cardiac outputs. Epinephrine, norepinephrine, reduction with pulmonary vasodilators, including inhaled
and dobutamine are classically infused to provide adrener- nitric oxide, prostaglandins, or phosphodiesterase
gic support to the systemic circulation. For patients with inhibitors; chronotropy achieved pharmacologically with
elevated central venous pressures, milrinone, a beta-agonists or with cardiac pacing, and mechanical cir-
phosphodiesterase-3 inhibitor, may be added to the inotro- culatory support using right VADs.
pic regimen for enhanced myocardial contractility and
reduction of right ventricular afterload due to its pulmo-
28.6.5 Primary Graft Failure
nary vasorelaxant properties. In cases of low systemic vas-
cular resistance, vasopressin or phenylephrine may Primary graft failure refers to a state of severe cardiac
provide necessary pressor support to maintain an adequate dysfunction resulting in profound circulatory insuffi-
perfusion pressure [42]. ciency within the first 24 h following transplantation. It is
The heart rate of the denervated heart is classically defined by a low cardiac output despite adequate filling
tachycardic. However, sinus node dysfunction attributed pressures, and may arise from either univentricular or
to graft ischemia, trauma, or inherent donor heart biventricular failure following exclusion of conditions
pathology may result in relative chronotropic incompe- with similar hemodynamic profiles including hyperacute
tence in the early postoperative period following trans- rejection and cardiac tamponade. Risk factors for the
plantation. Isoproterenol and theophylline have been development of primary graft failure can be attributed to
Chapter | 28 Current Status of Heart Transplantation 411

TABLE 28.3 ISHLT Acute Cellular Rejection Grades

Rejection Grade Description


None 0 Normal myocardium
Mild 1R # 1 focus of interstitial or perivascular
lymphocytic infiltration with myocyte
damage
Moderate 2R $ 2 foci of lymphocytic infiltration with
myocyte damage
Severe 3R Diffuse lymphocytic infiltration
Evidence of myocyte damage, edema,
hemorrhage, or vasculitis

FIGURE 28.3 Acute cellular rejection. Hematoxylin-eosin staining ISHLT grades of rejection (IR, 2R, and 3R) on the basis
demonstrating lymphocytic infiltration (thick arrow), and myocyte swell- of degree of lymphocytic infiltration and myocyte necro-
ing and necrosis (thin arrows). Image courtesy of Dr. Robert Padera,
Brigham and Women’s Hospital, Boston, MA.
sis (Figure 28.3; Table 28.3). However, recent ISHLT
registry data suggests that the threshold to treat rejection
appears to be rising with a 10% decline over a 5-year
donor, recipient, and perioperative conditions including period in the treatment of mild rejection occurring within
older age, inotropic requirements prior to surgery, renal the first year of transplant [56]. As such, treatment is typi-
failure, and graft ischemic time [54,55]. Primary graft cally reserved for moderate to severe rejection and may
failure is the main cause of 30 day mortality following consist of pulse corticosteroids, use of cytolytic therapy,
transplantation accounting for 39% of all deaths within or changes in calcineurin and antiproliferative exposure.
this period and nearly 20% within the first 12 months Antibody-mediated rejection (AMR) is characterized
[56]. Allograft management is supportive while efforts to by the adverse humoral response of preformed circulating
restore end-organ perfusion range from aggressive medi- antibodies directed against antigens within the cardiac
cal therapy using inotropes and vasopressors, to mechani- allograft that can occur days to weeks after transplantation.
cal circulatory support with intra-aortic balloon pumps, Risk factors include prior blood transfusions, multiparity,
extracorporeal membrane oxygenation (ECMO), or VADs pretransplant use of VADs, previous antibody exposure
to assist either left, right, or to both ventricles [57,58]. and retransplantation [59]. AMR is a significant risk factor
Cardiac recovery has been reported to occur within 24 h for poor long-term transplant outcomes, including sur-
and up to 7 days following hemodynamic support [58]. vival, graft failure, and the development of coronary allo-
graft vasculopathy [60,61]. A tissue diagnosis specifically
28.6.6 Hyperacute Rejection requiring immunostaining against C4D, which represents a
degradation product of activated complement C4b and is a
Hyperacute rejection represents the most severe of immu- marker of antibody activity (Figure 28.4). A mechanistic
nological reactions against the donor heart. Graft failure approach to the treatment of AMR targets the
arises within minutes to hours following transplantation. inactivation and elimination of circulating antibodies, the
Circulating antibodies within the recipient target the HLA suppression of T- and B-lymphocytes, plasma cell deple-
class I molecules within the donor heart vascular endothe- tion, and complement inhibition (Figure 28.5) [62].
lium. The consequent inflammatory response leads to
near immediate graft ischemia and necrosis. While
explantation of the donor heart is the only treatment 28.7 IMMUNOSUPPRESSION STRATEGIES
option, patient mortality is inevitable.
AND REJECTION SURVEILLANCE
Immunosuppressive therapy is required to abate the cell-
28.6.7 Acute Graft Rejection mediated and humoral immune responses that are directed
Acute cellular rejection is mediated by T-lymphocytes against the donor heart. Although a thorough elaboration
and can occur as early as within days of transplant, or as of transplant immunology and pharmacology extends
late as several years following. Diagnosis is made by tis- beyond the focus of this chapter, a guiding principle gov-
sue biopsy and can occur in up to 40% of patients within erning immunosuppression aims to optimally reduce the
the first year of transplant. Severity is assessed using incidence of transplant rejection without incurring the
412 PART | IV Heart

FIGURE 28.4 Antibody-


mediated rejection. Hematoxylin-
eosin staining (left) and C4D
staining (right). Images courtesy
of Dr. Robert Padera, Brigham
and Women’s Hospital, Boston,
MA.

Inhibition of circulating
antibodies

Intravenous immunoglobulin

T-cell suppression
Elimination of
circulating antibodies Calcineurin inhibitors
purine synthesis inhibitors
mTOR inhibitors
Plasmapheresis
IL-2 antibody
Plasma exchange
Antibody-mediated
rejection

(AMR)

Complement inhibition B-cell suppression

Eculizimab Rituximab
Splenectomy
Calcineurin inhibitors

Plasma cell depletion

Bortezomib

FIGURE 28.5 Targeted treatment of antibody-mediated rejection. Modified from Nair et al. [62].

untoward consequences of over-suppression, including of acute rejection. Such patients are known to have pre-
opportunistic infection and malignancy risk. Current formed circulating antibodies, including African
approaches to immunosuppression are guided by limited Americans, younger patients, and those with a history of
clinical trials and largely by institutional and transplant pregnancy, prior sternotomy, or mechanical circulatory
provider experience. support [59]. Alternatively, induction agents have been
used as an intermediary antirejection therapy in those
patients in whom renal insufficiency renders initiation of a
28.7.1 Induction Therapy calcineurin inhibitor prohibitive. Historically, induction
Induction therapy refers to the strategy in which an immu- therapy was implemented as a panacea to promote
nomodulating agent is administered to a highly sensitized immune tolerance; however, its contemporary use has
recipient immediately prior to transplant to reduce the risk remained controversial [63]. An observed incidence of
Chapter | 28 Current Status of Heart Transplantation 413

malignancy, including posttransplant lymphoproliferative immunosuppression on the basis of supportive clinical trial
disorder (PTLD) [64], in addition to cytomegalovirus and evidence: (i) corticosteroids, (ii) calcineurin inhibitors, (iii)
fungal infections [65,66] have limited its general use to antiproliferative agents. Mammalian target of rapamycin
approximately only half of all transplant centers [56]. (mTOR) inhibitors, also called proliferation signal inhibitors,
OKT3, a murine antithymoctye monoclonal antibody, represent a fourth and lesser utilized class of maintenance
targets the CD3 receptor of human T-cells rendering it immunosuppressive therapy.
vulnerable to opsonization and clearance by circulating
macrophages. However, its use has largely been elimi-
nated due to intolerable side effects, including a cytokine 28.8.1 Corticosteroids
release syndrome manifesting as fever, nausea, emesis,
Corticosteroids occupy a central role in the acute treat-
chest pain, and dyspnea [67], and its association with the
ment of rejection, as an induction agent and for chronic
delayed emergence of PTLD and opportunistic infections
maintenance immunosuppressive therapy. They provide
in a dose-dependent manner [68]. More commonly, poly-
broad immunosuppression by inhibiting the transcription
clonal antithymocyte antibodies, including the horse-
of multiple mediators of the posttransplant inflammatory
derived ATGAM and rabbit-based ATG, are implemented
cascade including IL-1, IL-2, CD40 ligand, TNF-alpha,
as induction agents due to a greater potency in reducing
gamma interferon, GM-CSF, and related growth factors.
T-lymphocytes without increasing long-term PTLD
However, concern with side effects, including psychosis,
malignancy risk [69,70].
cushingoid features, hypertension, avascular necrosis,
IL-2 receptor antagonists, including basiliximab and
dyslipidemia, steroid-induced hyperglycemia, and adrenal
dacluzimab, also confer immune tolerance by the deple-
insufficiency have led to the consideration of steroid-free
tion of circulating T-lymphocytes. Basiliximab, a
maintenance immunosuppression regimens. Adult and
chimeric human/mouse antibody, has been shown to be
pediatric studies suggest that overall rates of survival and
well tolerated as compared to placebo in de novo trans-
rejection are similar to those on corticosteroids [76,77].
plant patients treated concomitantly with cyclosporine,
However, as in the case of all immunosuppression strate-
mycophenolate mofetil (MMF), and steroids when admin-
gies, withdrawal of steroids is ultimately patient-specific
istered on Day 0 and Day 4 posttransplant [71]. When
depending on individual rejection history and chronicity
compared to OKT3 in the randomized multicenter com-
of steroid use. Current trends demonstrate that steroid use
parison of Basiliximab and Muromonab (OKT3) in heart
has gradually declined further out from transplantation
transplantation (SIMCOR) trial, basiliximab was found to
with more than 50% of patients being steroid-free 5 years
have greater tolerability with similar rates of infection,
posttransplantation [56]. Whether steroid-free immuno-
severity of rejection, and actuarial survival [72]. In com-
suppression alters late complications remains uncertain
parison to polyclonal antibody therapy with antithymo-
since low, relatively physiologic, doses are typically used
cyte globulin, additional studies have found basiliximab
in long-term survivors that continue maintenance steroids.
to have similar efficacy in composite endpoint and fre-
quency of acute rejection (Grade $ 1R), and better safety
endpoints including infectious death [73]. However, such
results have been contradicted by other findings suggest-
28.8.2 Calcineurin Inhibitors
ing that while ATG may have greater infectious risk, Cyclosporine and tacrolimus inhibit the enzymatic action
basiliximab was not a superior prophylactic against acute of calcineurin which plays an important role in the pro-
rejection [74]. duction of multiple cytokines including IL-2. In so doing,
The induction benefit of dacluzimab was studied in a calcineurin inhibitors limit T-cell activation and prolifera-
placebo-controlled randomized trial involving 434 new tion, a fundamental tenet of an immunosuppression proto-
transplant patients who were concomitantly treated with col. Cyclosporine had once been the de facto calcineurin
cyclosporine, MMF, and corticosteroids [75]. Despite a inhibitor; however, its use has largely been supplanted by
reduction in acute cellular rejection, the increased rate of tacrolimus [56]. Although studies have demonstrated sim-
infection-related mortality signaled the death of dacluzi- ilar 1 year survival between cyclosporine and tacrolimus
mab, leading to its subsequent withdrawal from market [78,79], the risk of acute rejection was found to be lower
production. in one clinical trial [80] and further supported by a recent
meta-analysis [81]. Despite similar efficacy, tacrolimus
28.8 MAINTENANCE has a less adverse complication profile favoring its
current use. Cyclosporine use can be complicated by
IMMUNOSUPPRESSION hypertension, cholelithiasis, dyslipidemia, and gingival
Three classes of medications are typically instituted hyperplasia [78 80], in addition to significant drug inter-
early after cardiac transplantation for maintenance actions. Moreover, the results of the TICTAC trial have
414 PART | IV Heart

reinforced the use of tacrolimus as a pivotal agent in any of a maintenance immunosuppression regimen for those
immunosuppression regimen. In this trial, tacrolimus patients with transplant-related vasculopathy. However,
monotherapy resulted in a 6 and 12 month freedom from general mTOR inhibitor use has been limited by their
severe cellular rejection (ISHLT Grade $ 2) of 93.3% as association with poor wound healing, renal failure, dysli-
compared to 92.9% in the tacrolimus/mycophenolate pidemia, anemia, thrombocytopenia, and the development
combination group [82]. In addition, the 3-arm trial com- of both pleural and pericardial effusions [88].
paring combination therapies of tacrolimus/MMF versus
tacrolimus/sirolimus versus cyclosporine/MMF found a
lower incidence of treated rejection among patients trea-
28.9 REJECTION SURVEILLANCE
ted with tacrolimus [83]. However, tacrolimus can Acute rejection has been reported to occur in 24.5% of
increase the risk of diabetes and induce neurological com- patients within the first year following transplant. Within
plications. In patients with either chronic renal insuffi- 5 years, 50.9% of all transplant patients experience at
ciency or acute kidney failure posttransplant, delayed least one episode of acute rejection [34]. Therefore, each
tacrolimus initiation may be necessary and calcineurin institution has its own schedule and methodology for
inhibitor-based immunosuppression preempted by induc- rejection monitoring. Biopsy-proven rejection, particu-
tion therapy. larly those categorized as mild in severity, largely occurs
in the absence of symptoms or even allograft dysfunction.
Rejection may present clinically across a spectrum to also
28.8.3 Antiproliferative Agents include new onset brady- or tachyarrhythmias manifesting
Antiproliferative agents, also known as antimetabolites, as palpitations or more significantly as syncope, the
inhibit cell-cycle pathways to limit T- and B-cell prolifer- reemergence of heart failure symptoms due to a failing
ation and thereby reducing the cytotoxic response directed graft, or even sudden cardiac death [90]. Symptom-
toward the cardiac allograft. Azathioprine, the original rejection mismatch consequently results in measures of
antiproliferative drug, mediates its effects through a routine surveillance requiring the transplant patient to
purine analog metabolite that arrests DNA synthesis once undergo a potential battery of testing modalities to moni-
incorporated into the lymphocytic nuclei. MMF and tor for cardiac allograft rejection.
mycophenolic acid are reversible inhibitors of the enzyme
inosine monophosphate dehydrogenase. This enzyme
serves as an important immunosuppression target as it is
28.9.1 Endomyocardial Biopsy
both upregulated during T- and B-cell activation and Historically, noninvasive markers, including cardiac
forms part of an obligate pathway in purine synthesis nec- enzymes or low QRS voltage on surface electrocardio-
essary for their proliferation [84]. Contemporary practice gram due to myocardial edema or fibrosis, have been
trends highlight greater utilization of MMF and myoco- used to signal subclinical rejection. Endomyocardial
phenolic acid over azathioprine for maintenance immuno- biopsy (EMB) remains the contemporary gold standard
suppressive therapy [56]. In comparison to azathioprine, for monitoring of graft rejection. Tissue samples are
MMF promotes greater survival, less rejection, and less obtained from the right ventricular septum using a biop-
coronary allograft vasculopathy posttransplant [85 87]. tome introduced through the right internal jugular vein,
Azathioprine and MMF both can induce significant leuko- typically under fluoroscopy or by echocardiographic guid-
penia from profound bone marrow suppression. MMF can ance [91]. The additional use of a right heart catheter
induce significant gastrointestinal difficulties including facilitates the measurement of intracardiac pressures and
diarrhea, gastritis, and nausea. cardiac output. As an invasive procedure, EMBs carry a
complication risk of 0.5% in some reports [92,93].
Complications may arise from central venous access
28.8.4 mTOR Inhibitors including inadvertent arterial puncture, bleeding, and
mTOR is an enzyme kinase that mediates lymphocyte pneumothorax. Alternatively, injury may result from the
growth, differentiation, and proliferation. Sirolimus and biopsy itself including cardiac perforation leading to tam-
everolimus are mTOR inhibitors that in addition to their ponade, arrhythmia, and accidental sampling of tricuspid
effects on T- and B-cells also inhibit the proliferation of valve leaflets leading to significant valvular regurgitation
the vascular smooth muscle wall which has broader clini- [94]. Tissue biopsy yields invaluable information regard-
cal significance for the treatment of cardiac allograft ing the presence of rejection, type (cellular versus anti-
vasculopathy (CAV). In comparison to azathioprine, sirio- body), severity, and chronicity, in addition to pathological
limus and everolimus have been shown to have less rejec- tissue changes suggestive of CAV. While institutional
tion and a lower burden of coronary allograft practice may vary, typical biopsy schedules include
vasculopathy [88,89]. Sirolimus may be included as part EMBs performed weekly for the first 4 weeks with a
Chapter | 28 Current Status of Heart Transplantation 415

tapered frequency over the course of the first 12 months etiologies of the highest survivorship. In 2011, the rate of
following transplant. Beyond the first year, EMBs are graft failure within the first 6 weeks posttransplant
performed annually, or as motivated by patient symptoms. improved to 4.9% as compared to previous reports [34].
Some programs advocate cessation of routine biopsy sur- Risk factors for death within the first year include graft
veillance after the first 2 years of transplant with similar ischemic times greater than 200 min, extremes of recipi-
reported outcomes to those of continued intensive ent age, and a pretransplant requirement for mechanical
surveillance. circulatory support. When stratified according to the first
3 years posttransplant, causes for early death are predomi-
nantly associated with graft failure and overwhelming
28.9.2 Biomarker Tests infection. Beyond 3 years, mortality is attributed to malig-
Gene expression profiling is an alternative noninvasive nancy and the development of CAV [56]. Overall graft
strategy to assess for the absence of acute cellular rejec- survival at 5 years approximates 74.9% regardless of
tion [95 97]. With one such technique, Allomap (XDS), medical urgency (Status 1A, 1B, or 2) or underlying heart
RNA from peripheral blood mononuclear cells is isolated failure etiology [34]. For those patients surviving their
and amplified using polymerase chain reaction (PCR) first year posttransplant, data from the Scientific Registry
techniques to facilitate gene expression profiles of 11 of Transplant Recipients suggest that their predicted half-
genes known to distinguish between rejection and nonre- life is 14.0 years. In fact, both graft and patient outcomes
jection in posttransplant patients. A score ranging from 0 have continued to improve with 21,457 survivors living
to 40 is tabulated using an algorithm based on candidate in 2011 as compared to 16,259 in 2001 [34], with improv-
gene expression levels. A score less than 30 has a nega- ing trends over the last three decades (Figure 28.6).
tive predictive value for biopsy-proven cellular rejection Posttransplant morbidity is typically associated with
of 99.6% [95] and is deemed to obviate the need for an renal failure, malignancy, acute rejection, and CAV [56].
EMB. Although promising, the technique has been lim- Patients have an increasing risk of renal failure over time
ited by a lack of high specificity and the inability to dis- following transplant with an incidence of 6%, 11%, and
tinguish between cellular and antibody-mediated rejection 16% at 1, 3, and 5 years posttransplant, respectively. Risk
[98]. In the IMAGE trial [95], the use of this gene expres- factors include pretransplant recipient comorbidities of
sion profiling biomarker was shown to decrease the num- increasing age, history of diabetes, and the preexistence
ber of biopsies required in surveillance while maintaining of an elevated serum creatinine. Within the first year
clinical outcomes. However, some have argued that the posttransplant, mortality is typically attributed to acute
gold standard of surveillance biopsies beyond 5 months rejection, infection, and graft failure. At 5 and 10 years
may itself be a flawed technique and appropriate compari- posttransplant, the mortality burden arises from malig-
son of the noninvasive biomarker ought to rest on its nancy contributing to over 20% of deaths with an inci-
incremental value to clinical and allograft functional dence of 6% and 15%, respectively (Figure 28.7). Skin
based monitoring. The cost-effectiveness of this approach cancer is the most common malignancy in the transplant
remains uncertain as does its overall routine use in clini- patient. Other malignancies include lymphoproliferative
cal practice [99]. disorders and cancers of the prostate, lung, bladder, kid-
ImmuKnow (Cylex Inc.) is an immune function assay ney, breast, and colon. Risk factors include recipient age
which tests the ATP production from a transplant and long-term immunosuppression [42].
patient’s isolated T-lymphocytes [100]. By exposing these
cells to a T-cell mitogen, plant phytohemaglutinin, mea-
sured ATP can identify those patients who may be either
28.10.1 Cardiac Allograft Vasculopathy
excessively or inadequately immunosuppressed. In addi- CAV remains the most indolent long-term complication
tion to tailoring immunosuppression, the assay may be affecting the donor heart coronary vasculature following
able to predict patients at high risk for graft rejection. transplantation. Unlike an atherosclerotic plaque, CAV is
However, its adoption into mainstream transplant practice classically characterized by the diffuse and progressive
is pending larger-scale clinical trial data. Current observa- proliferation of the intimal layer of the arterial wall and
tional evidence suggests that its ability to predict infec- can extend from epicardial vessels to deep penetrating
tion may be greater than its ability to inform on rejection. resistance arterioles and capillaries (Figure 28.8).
However, the remodeling process involves all three layers
of the vascular wall and is influenced by adventitial scar-
28.10 TRANSPLANTATION OUTCOMES ring and alterations in medial tone. These hyperplastic
One year life expectancy following heart transplant lesions can lead to luminal narrowing sufficient to
ranges from 79% to 86% depending on the underlying obstruct coronary blood flow. Although episodes of cellu-
cardiomyopathy of the recipient, with nonischemic lar and antibody-mediated rejection have been associated
416 PART | IV Heart

100
1982–1992vs. 1993–2002: p < 0.0001
1982–1992vs. 2003–6/2002: p < 0.0001
1993–2002vs. 2003–6/2010: p < 0.0001
80
Survival (%)

60

1982–1992 (N = 25, 138)


1993–2002 (N = 37, 193)
40
2003–6/2010 (N = 24, 021)

20

Median survival: 1982–1992: 8.5 years: 1993–2002: 10.9 years: 2003–6/20120: NA


0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15
Years

FIGURE 28.6 Cardiac transplant survival curves according to transplant era. Figure reprinted from [33].

100
Acute rejection CAV Graft failure
90 Renal failure Infection (Non-CMV) Malignancy
80

70 11
20
23 23
% of deaths

60 3
0 14
50 2 11
13
29 12 11
40 0 4
27 6 9
30 1 25
34 18 16
20 17
10
10 3 10
12 11 13
1 9
4 5 2 1
0
0–30 days 31 days–1 year >1 year – 3 years >3 year – 5 years >5 year –10 >10 Years
(N=1817) (N=1750) (N=1245) (N=1112) years (N=3321) (N=5035)

FIGURE 28.7 Causes of death posttransplant. Figure reprinted from [33].

with CAV, current immunosuppression protocols are lim- cytokines, including interferon-γ and the cellular agents
ited in their ability to prevent or delay its of innate immunity, such as T-cells, macrophages, and
inevitable occurrence. Numerous risk factors have been monocytes. Although 10% of patients can develop CAV
associated with the development of CAV including age, as early as within the first year of transplant, it is as much
sex, preexisting coronary artery disease, and cytomegalo- a marker of graft survival with a prevalence of greater
virus infection in both donor and recipient. Most impor- than 50% in those patients surviving at least 10 years fol-
tantly, the number and severity of acute cellular rejection lowing transplantation [56].
predisposes to the development of CAV suggesting that Coronary angiography remains the gold standard for
its predisposition is more likely a manifestation of the the evaluation of CAV. Severity of CAV is classified
immunogenicity of the cardiac allograft itself. according to the degree of luminal narrowing of the
Mechanistic studies demonstrate that CAV is the inflam- affected vessel (e.g., left main), its associated anatomic
matory consequence of a vascular remodeling process location (proximal versus distal third), and the total num-
mediated by the complex interplay between numerous ber of vessels involved [60]. Greater severity is ascribed
Chapter | 28 Current Status of Heart Transplantation 417

FIGURE 28.8 CAV—gross tis-


sue specimen (left) demonstrating
significant CAV-related vascular
narrowing of a coronary artery (*)
as compared to an unaffected adja-
cent larger vessel. Intimal prolifer-
ation with near luminal obliteration
(arrow) by hematoxylin-eosin
staining (right). Images courtesy of
Dr. Robert Padera, Brigham and
Women’s Hospital, Boston, MA.

to those lesions associated with allograft dysfunction. importantly, revascularization does not reduce the burden
This may be the result of systolic impairment with a of allograft rejection which is the predominant risk
measured ejection fraction of less than 45% or with factor for CAV progression [105,106]. As such, retrans-
hemodynamic or echocardiographic indices of restrictive plantation remains the definitive treatment for severe
physiology, if contractility is preserved. Other imaging CAV in those patients deemed eligible.
modalities that have been employed to evaluate include
intravascular ultrasound (IVUS), myocardial perfusion
scans including technetium-99m sestamibi, dobutamine 28.11 MECHANICAL HORIZON: VADS AS
stress echocardiography, and multidetector computed
tomography.
A BRIDGE TO TRANSPLANT
CAV prevention includes the use of statins predomi- VADs provide mechanical circulatory support to the fail-
nantly for their lipid lower benefit. Small prospective ing heart. Since the initial use of the first artificial heart
clinical trials using pravastatin and simvastatin demon- device in 1963 for postcardiotomy shock [107], this tech-
strated long-term benefits in the reduction of CAV, inti- nology has evolved to significantly alter the therapeutic
mal thickness, and improved patient survival [101 103]. options for patients with end-stage cardiomyopathy.
The early initiation of diltiazem, often used to treat corti- There has been a proliferation of their use, with over
costeroid or cyclosporine-induced hypertension, was also 6000 VADs implanted in the United States [108].
found to be beneficial in CAV prevention [104]. Technological advancements over the last five decades
However, these studies were conducted in the prestatin has led to the development of smaller VADs that can pro-
era of transplant management. An adjuvant approach to vide either pulsatile or continuous blood flow to the left,
immunosuppression in patients with documented CAV right, or both ventricles.
consists of the antiproliferative drugs, everolimus and sir- VADs have been implanted in patients for any of four
olimus. In addition to decreasing rejection events, these indications. As destination therapy (DT), VADs provide
drugs have also been shown to reduce CAV [88,89] as long-term hemodynamic support for patients who are
measured by IVUS detection of intimal thickening. deemed transplant ineligible. Bridge to recovery (BTR)
However, issues with poor wound healing, renal failure, refers to the strategy by which VADs are implanted in
marrow suppression, and drug interaction have limited patients with significant cardiac decompensation but antic-
their use, and specifically for everolimus, FDA approval. ipated myocardial recovery, such as postcardiotomy
It is unclear whether the reduction in surrogate parameters shock, peripartum cardiomyopathy, or fulminant myocar-
of intimal thickening will indeed be accompanied by ditis. For transplant candidates awaiting heart replacement,
reduced cardiac allograft adverse events. VADs are used as a bridge to transplantation (BTT).
Revascularization is often limited due to the diffuse- Bridge to candidacy (BTC) represents a growing popula-
ness of disease along the coronary vasculature. tion of patients implanted with VADs who, although ini-
Angioplasty by percutaneous intervention may be an tially deemed transplant ineligible, are afforded both the
option for patients with single-vessel disease. However, benefits of time and augmented perfusion to enable a
stent patency may be at risk to restenosis by the same reconsideration of their transplant status.
immune factors directed against the heart. Coronary Initial US FDA approval for BTT VADs took place in
bypass surgery is typically limited due to the lack of via- 1994 with older generation devices. Since 2006, data
ble distal vessel targets due to intimal proliferation. Most from the Interagency Registry for Mechanically Assisted
418 PART | IV Heart

FIGURE 28.9 Use of mechanical


circulatory support (biventricular and
univentricular devices) as a “bridge”
to cardiac transplant. Figure reprinted
from [33].

FIGURE 28.10 VADs currently


FDA approved as a bridge to cardiac
transplantation: Thoratec HeartMate
II LVAD (Pleasanton, CA; left);
HeartWare VAD (Framingham,
MA; middle); Syncardia Systems
Total Artificial Heart (Tuscon, AZ;
right).

Circulatory Support (INTERMACS) demonstrate that at function. Unfortunately, such devices are inadequate in
least 1600 VADs were implanted over a 5 year period of patients with biventricular failure. The total artificial heart
time (2006 2011) as a bridge to transplant strategy. (TAH, Syncardia) provides biventricular mechanical sup-
ISHLT registry data also reveal that 36% of patients port. It is true heart replacement therapy as the surgery
transplanted in 2010 were supported with mechanical cir- requires excision of both right and left ventricles such
culatory devices reflecting a rising trend in transplant can- that the device can be anastomosed to both atria. In its
didates with BTT VADs [109] (Figure 28.9). clinical trial, patients with the TAH had a 70% 1-year
The predominant contemporary VAD in use is the survival rate and a 79% survival to transplant [111,112]
HeartMate II (Thoratec, Pleasanton, CA) which provides (Figure 28.10).
left ventricular support for patients for either DT or as a Despite clinical benefits and improved survivorship
BTT. It gained FDA approval in 2008 after pivotal clini- while waiting for transplant, VAD-related complications
cal trial findings demonstrating improved symptoms sta- including device failure, infection, and cerebrovascular
tus, functional capacity, and a 68% 1-year survival rate events due to embolic strokes or hemorrhage may com-
following implantation in patients awaiting a heart trans- promise transplant candidacy. In addition, device mainte-
plant [110]. More recently, the Heartware Ventricular nance requires anticoagulation which can further
Assist Device (HVAD, Framingham, MA) was granted compound bleeding diatheses that can result from an
FDA approval as a BTT strategy following results of the acquired von Willebrand disease and neovascular forma-
ADVANCE trial that demonstrated an 86% 1-year sur- tions that arise as consequences of nonpulsatile VAD
vival rate with improved quality of life parameters in flow [113,114]. Excessive blood loss may require transfu-
patients awaiting transplant [26]. Smaller than the sions that increase sensitization risks and consequent
HeartMate II, the HVAD can be surgically implanted allograft rejection or failure [115]. Impact of LVAD ther-
within the intrapericardial space to provide left ventricu- apy on transplant survival has been controversial
lar support. However, as left ventricular support devices, [110,116]. Recent data, however, suggest patients, previ-
both require a relative preservation of right ventricular ously supported with continuous flow VADs, who survive
Chapter | 28 Current Status of Heart Transplantation 419

beyond the initial 6 months posttransplant, have similar based therapies hold greater promise for those with end-
long-term survival outcomes as compared to conventional stage heart disease.
transplantation [56,117].
The era of mechanical circulatory support has also
afforded those patients with DT devices, the opportunity ACKNOWLEDGMENTS
for transplant reconsideration. At least 10% patients with
We express our sincere gratitude to Dr. Robert Padera for contribut-
DT VADs, originally intended a palliative therapy, were ing pathological figures to supplement this manuscript.
transplanted within 12 months following implantation
[109]. In fact, the benefits of improved hemodynamics
and organ perfusion by mechanical circulatory support
REFERENCES
have rendered the distinction between absolute and rela-
tive contraindication less dichotomous. Traditional exclu- [1] Hunt SA, Abraham WT, Chin MH, Feldman AM, Francis GS,
sion criteria, such as renal failure and pulmonary Ganiats TG, et al. Focused update incorporated into the ACC/
hypertension, have been found to be “VAD-modifiable” AHA 2005 guidelines for the diagnosis and management of heart
failure in adults: a report of the American College of Cardiology
due to mechanical unloading and improved cardiac output
Foundation/American Heart Association Task Force on Practice
[118]. Improved survival with the VAD also affords valu-
Guidelines: developed in collaboration with the International
able time for reevaluation of other modifiable contraindi- Society for Heart and Lung Transplantation. Circulation
cations including smoking, illicit drug and alcohol abuse, 2009;119:391 479.
BMI, and patient attitude toward transplant. To this end, [2] Nohria A, Tsang SW, Fang JC, Lewis EF, Jarcho JA, Mudge GH,
from 2006 to 2011, 1765 VADs were implanted in et al. Clinical assessment identifies hemodynamic profiles that
patients as a BTC allowing more individuals the opportu- predict outcomes in patients admitted with heart failure. J Am
nity for a new heart [109]. Coll Cardiol 2003;41:1797 804.
[3] Drazner MH, Hellkamp AS, Leier CV, Shah MR, Miller LW,
Russell SD, et al. Value of clinician assessment of hemodynamics
28.12 FUTURE DIRECTIONS IN CARDIAC in advanced heart failure: the ESCAPE trial. Circ Heart Fail
2008;1:170 7.
TRANSPLANTATION [4] Thomas SS, Nohria A. Hemodynamic classifications of acute
In 2010, the National Heart, Lung, and Blood Institute heart failure and their clinical application: an update. Circ J
Working Group convened to outline research strategies 2012;76:278 86.
that will ultimately improve the clinical decision-making [5] Pfeffer MA, Lamas GA, Vaughan DE, Parisi AF, Braunwald E.
Effect of captopril on progressive ventricular dilatation after ante-
required in the evolving care of cardiac transplant patients
rior myocardial infarction. N Engl J Med 1988;319:80 6.
[119]. Over the next 10 years, research efforts will be
[6] Arnold JM, Yusuf S, Young J, Mathew J, Johnstone D, Avezum
encouraged to improve long-term allograft outcomes, fos- A, et al. Prevention of heart failure in patients in the Heart
ter tailored immunotherapy, and expand the available Outcomes Prevention Evaluation (HOPE) study. Circulation
donor pool through improved graft preservation strategies 2003;107:1284 90.
and marginal candidate optimization. In addition, [7] Flather MD, Yusuf S, Kober L, Pfeffer M, Hall A, Murray G,
advancements in the field will require robust clinical et al. Long-term ACE-inhibitor therapy in patients with heart
trials that explore mechanisms and treatment of humoral failure or left-ventricular dysfunction: a systematic overview of
and cellular-mediated rejection. This is in addition to the data from individual patients. ACE-Inhibitor myocardial infarc-
need for technological advances in both mechanical circu- tion collaborative group. Lancet 2000;355:1575 81.
latory support and stem cell therapies that may promote [8] Granger CB, McMurray JJ, Yusuf S, Held P, Michelson EL,
Olofsson B, et al. Effects of candesartan in patients with chronic
true cardiac regeneration.
heart failure and reduced left-ventricular systolic function intoler-
ant to angiotensin-converting-enzyme inhibitors: the CHARM—
Alternative trial. Lancet 2003;362:772 6.
28.13 CONCLUSION [9] Granger CB, McMurray JJ, Yusuf S, Held P, Michelson EL,
The management of end-stage heart disease has evolved Olofsson B, et al. Effects of candesartan in patients with chronic
heart failure and reduced left-ventricular systolic function taking
with our improved understanding of its complex patho-
angiotensin-converting-enzyme inhibitors: the CHARM—Added
physiology. Since the first heart transplant in 1969, the
trial. Lancet 2003;362:767 71.
field of cardiac transplant medicine continues to evolve.
[10] Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJ,
In the end, however, the promise of heart replacement Michelson EL, et al. Effects of candesartan on mortality and mor-
therapy is limited foremost by its availability. Until such bidity in patients with chronic heart failure: the CHARM—
time as more effective global donor promotion strategies Overall programme. Lancet 2003;362:759 66.
are in place, our sights must rest upon a horizon in which [11] Yusuf S, Pfeffer MA, Swedberg K, Granger CB, Held P,
mechanical circulatory support devices and stem cell- McMurray JJ, et al. Effects of candesartan in patients with chronic
420 PART | IV Heart

heart failure and preserved left-ventricular ejection fraction: the [27] Slaughter MS, Rogers JG, Milano CA, Russell SD, Conte JV,
CHARM—Preserved Trial. Lancet 2003;362:777 81. Feldman D, et al. Advanced heart failure treated with continuous-
[12] Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, et flow left ventricular assist device. N Engl J Med
al. The effect of spironolactone on morbidity and mortality in 2009;361:2241 51.
patients with severe heart failure. Randomized aldactone evalua- [28] Barnard CN. The operation. A human cardiac transplant: an
tion study investigators. N Engl J Med 1999;341:709 17. interim report of a successful operation performed at Groote
[13] Zannad F, McMurray JJ, Krum H, van Veldhuisen DJ, Swedberg Schuur Hospital, Cape Town. S Afr Med J 1967;41:1271 4.
K, Shi H, et al. Eplerenone in patients with systolic heart failure [29] Sade RM. Transplantation at 100 years: Alexis Carrel, pioneer
and mild symptoms. N Engl J Med 2011;364:11 21. surgeon. Ann Thorac Surg 2005;80:2415 8.
[14] Packer M, Coats AJ, Fowler MB, Katus HA, Krum H, Mohacsi P, [30] Hardy JD, Kurrus FD, Chavez CM, Neely WA, Eraslan S, Turner
et al. Effect of carvedilol on survival in severe chronic heart fail- MD, et al. Heart transplantation in man. Developmental studies
ure. N Engl J Med 2001;344:1651 8. and report of a case. JAMA 1964;188:1132 40.
[15] The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a rando- [31] Caves PK, Stinson EB, Billingham ME, Rider AK, Shumway NE.
mised trial. Lancet 1999;353(9146):9 13. Diagnosis of human cardiac allograft rejection by serial cardiac
[16] Moss AJ, Zareba W, Hall WJ, Klein H, Wilber DJ, Cannom DS, biopsy. J Thorac Cardiovasc Surg 1973;66:461 6.
et al. Prophylactic implantation of a defibrillator in patients with [32] Modry DL, Oyer PE, Jamieson SW, Stinson EB, Baldwin JC,
myocardial infarction and reduced ejection fraction. N Engl J Reitz BA, et al. Cyclosporine in heart and heart lung transplanta-
Med 2002;346:877 83. tion. Can J Surg 1985;28:274 80.
[17] Solomon SD, Foster E, Bourgoun M, Shah A, Viloria E, Brown [33] Stehlik J, Edwards LB, Kucheryavaya AY, Benden C, Christie
MW, et al. Effect of cardiac resynchronization therapy on reverse JD, Dipchand AI, et al. The Registry of the International Society
remodeling and relation to outcome: multicenter automatic defi- for Heart and Lung Transplantation: 29th official adult
brillator implantation trial: cardiac resynchronization therapy. heart transplant report—2012. J Heart Lung Transplant
Circulation 2010;122:985 92. 2012;31:1052 64.
[18] Tang AS, Wells GA, Talajic M, Arnold MO, Sheldon R, [34] Colvin-Adams M, Smith JM, Heubner BM, Skeans MA, Edwards
Connolly S, et al. Cardiac-resynchronization therapy for mild-to- LB, Waller C, et al. OPTN/SRTR 2011 annual data report: heart.
moderate heart failure. N Engl J Med 2010;363:2385 95. Am J Transplant 2011;13:119 48.
[19] Felker GM, Thompson RE, Hare JM, Hruban RH, Clemetson DE, [35] Mancini DM, Eisen H, Kussmaul W, Mull R, Edmunds Jr. LH,
Howard DL, et al. Underlying causes and long-term survival in Wilson JR. Value of peak exercise oxygen consumption for opti-
patients with initially unexplained cardiomyopathy. N Engl J Med mal timing of cardiac transplantation in ambulatory patients with
2000;342:1077 84. heart failure. Circulation 1991;83:778 86.
[20] McCarthy 3rd RE, Boehmer JP, Hruban RH, Hutchins GM, [36] Elmariah S, Goldberg LR, Allen MT, Kao A. Effects of gender on
Kasper EK, Hare JM, et al. Long-term outcome of fulminant myo- peak oxygen consumption and the timing of cardiac transplanta-
carditis as compared with acute (nonfulminant) myocarditis. tion. J Am Coll Cardiol 2006;47:2237 42.
N Engl J Med 2000;342:690 5. [37] Elmariah S, Goldberg LR, Allen MT, Kao A. The effects of race
[21] Konstam MA, Kramer DG, Patel AR, Maron MS, Udelson JE. on peak oxygen consumption and survival in patients with systolic
Left ventricular remodeling in heart failure: current concepts in dysfunction. J Card Fail 2010;16:332 9.
clinical significance and assessment. JACC Cardiovasc Imaging [38] Peterson LR, Schechtman KB, Ewald GA, Geltman EM, de las
2011;4:98 108. Fuentes L, Meyer T, et al. Timing of cardiac transplantation in
[22] Renlund DG, Kfoury AG. When the failing, end-stage heart is not patients with heart failure receiving beta-adrenergic blockers.
end-stage. N Engl J Med 2006;355:1922 5. J Heart Lung Transplant 2003;22:1141 8.
[23] Mehra MR, Domanski MJ. Should left ventricular assist device [39] Goda A, Lund LH, Mancini D. The heart failure survival score
should be standard of care for patients with refractory heart failure outperforms the peak oxygen consumption for heart transplanta-
who are not transplantation candidates?: left ventricular assist tion selection in the era of device therapy. J Heart Lung
devices should be considered standard of care for patients with Transplant 2011;30:315 25.
refractory heart failure who are not transplantation candidates. [40] Mancini D, Lietz K. Selection of cardiac transplantation candi-
Circulation 2012;126:3081 7. dates in 2010. Circulation 2010;122:173 83.
[24] Cuffe MS, Califf RM, Adams Jr. KF, Benza R, Bourge R, [41] Mehra MR, Kobashigawa J, Starling R, Russell S, Uber PA,
Colucci WS, et al. Short-term intravenous milrinone for acute Parameshwar J, et al. Listing criteria for heart transplantation:
exacerbation of chronic heart failure: a randomized controlled international society for heart and lung transplantation guidelines
trial. JAMA 2002;287:1541 7. for the care of cardiac transplant candidates 2006. J Heart Lung
[25] Bayram M, De Luca L, Massie MB, Gheorghiade M. Transplant 2006;25:1024 42.
Reassessment of dobutamine, dopamine, and milrinone in the [42] Costanzo MR, Dipchand A, Starling R, Anderson A, Chan M,
management of acute heart failure syndromes. Am J Cardiol Desai S, et al. The International Society of Heart And Lung
2005;96:47 58. Transplantation guidelines for the care of heart transplant recipi-
[26] Aaronson KD, Slaughter MS, Miller LW, McGee EC, Cotts WG, ents. J Heart Lung Transplant 2010;29:914 56.
Acker MA, et al. Use of an intrapericardial, continuous-flow, cen- [43] Uriel N, Jorde UP, Cotarlan V, Colombo PC, Farr M, Restaino
trifugal pump in patients awaiting heart transplantation. SW, et al. Heart transplantation in human immunodeficiency
Circulation 2012;125:3191 200. virus-positive patients. J Heart Lung Transplant 2009;28:667 9.
Chapter | 28 Current Status of Heart Transplantation 421

[44] Qiu J, Terasaki PI, Waki K, Cai J, Gjertson DW. HIV-positive early cardiac allograft dysfunction. J Heart Lung Transplant
renal recipients can achieve survival rates similar to those of 2011;30(7):783 9; PubMed PMID: 21481606.
HIV-negative patients. Transplantation 2006;81:1658 61. [59] Mehra MR, Uber PA, Uber WE, Scott RL, Park MH.
[45] Hosenpud JD, Pamidi SR, Fiol BS, Cinquegrani MP, Keck BM. Allosensitization in heart transplantation: implications and man-
Outcomes in patients who are hepatitis B surface antigen-positive agement strategies. Curr Opin Cardiol 2003;18(2):153 8;
before transplantation: an analysis and study using the joint PubMed PMID: 12652223.
ISHLT/UNOS thoracic registry. J Heart Lung Transplant [60] Berry GJ, Angelini A, Burke MM, Bruneval P, Fishbein MC,
2000;19:781 5. Hammond E, et al. The ISHLT working formulation for patho-
[46] Lin MH, Chou NK, Chi NH, Chen YS, Yu HY, Huang SC, et al. logic diagnosis of antibody-mediated rejection in heart transplan-
The outcome of heart transplantation in hepatitis C-positive reci- tation: evolution and current status (2005 2011). J Heart Lung
pients. Transplant Proc 2012;44:890 3. Transplant 2011;30(6):601 11; PubMed PMID: 21555100.
[47] Corbett C, Armstrong MJ, Neuberger J. Tobacco smoking and [61] Prada-Delgado O, Estevez-Loureiro R, Paniagua-Martin MJ, Lopez-
solid organ transplantation. Transplantation 2012;94:979 87. Sainz A, Crespo-Leiro MG. Prevalence and prognostic value of car-
[48] Psychosocial Outcomes Workgroup of the N, Social Sciences diac allograft vasculopathy 1 year after heart transplantation accord-
Council of the International Society for H, Lung T, Cupples S, ing to the ISHLT recommended nomenclature. J Heart Lung
Dew MA, Grady KL, et al. Report of the psychosocial outcomes Transplant 2012;31(3):332 3; PubMed PMID: 22333404.
workgroup of the nursing and social sciences council of the inter- [62] Nair N, Ball T, Uber PA, Mehra MR. Current and future chal-
national society for heart and lung transplantation: present status lenges in therapy for antibody-mediated rejection. J Heart Lung
of research on psychosocial outcomes in cardiothoracic transplan- Transplant 2011;30(6):612 7; PubMed PMID: 21474341.
tation: review and recommendations for the field. J Heart Lung [63] Uber PA, Mehra MR. Induction therapy in heart transplantation:
Transplant 2006;25:716 25. is there a role? J Heart Lung Transplant 2007;26(3):205 9;
[49] Cohen O, De La Zerda DJ, Beygui R, Hekmat D, Laks H. Donor PubMed PMID: 17346621.
brain death mechanisms and outcomes after heart transplantation. [64] Swinnen LJ, Costanzo-Nordin MR, Fisher SG, O’Sullivan EJ,
Transplant Proc 2007;39:2964 9. Johnson MR, Heroux AL, et al. Increased incidence of lympho-
[50] Karamlou T, Shen I, Slater M, Crispell K, Chan B, Ravichandran proliferative disorder after immunosuppression with the monoclo-
P. Decreased recipient survival following orthotopic heart trans- nal antibody OKT3 in cardiac-transplant recipients. N Engl J Med
plantation with use of hearts from donors with projectile brain 1990;323(25):1723 8; PubMed PMID: 2100991.
injury. J Heart Lung Transplant 2005;24:29 33. [65] Costanzo-Nordin MR, Swinnen LJ, Fisher SG, O’Sullivan EJ,
[51] Swanson DK, Pasaoglu I, Berkoff HA, Southard JA, Hegge JO. Pifarre R, Heroux AL, et al. Cytomegalovirus infections in heart
Improved heart preservation with UW preservation solution. J transplant recipients: relationship to immunosuppression. J Heart
Heart Transplant 1988;7:456 67. Lung Transplant 1992;11(5):837 46; PubMed PMID: 1329959.
[52] Braunwald E. Innervation of the transplanted heart. N Engl J Med [66] Johnson MR, Mullen GM, O’Sullivan EJ, Liao Y, Heroux AL,
1969;281(15):848 9; PubMed PMID: 4897379. Kao WG, et al. Risk/benefit ratio of perioperative OKT3 in car-
[53] Mohanty PK, Sowers JR, Thames MD, Beck FW, Kawaguchi A, diac transplantation. Am J Cardiol 1994;74(3):261 6; PubMed
Lower RR. Myocardial norepinephrine, epinephrine and dopamine PMID: 8037132.
concentrations after cardiac autotransplantation in dogs. J Am [67] Lindenfeld J, Miller GG, Shakar SF, Zolty R, Lowes BD, Wolfel
Coll Cardiol 1986;7(2):419 24; PubMed PMID: 3511122. EE, et al. Drug therapy in the heart transplant recipient: part I:
[54] Kilic A, Weiss ES, Arnaoutakis GJ, George TJ, Conte JV, Shah cardiac rejection and immunosuppressive drugs. Circulation
AS, et al. Identifying recipients at high risk for graft failure after 2004;110(24):3734 40; PubMed PMID: 15596559.
heart retransplantation. Ann Thorac Surg 2012;93(3):712 6; [68] Frist WH, Merrill WH, Eastburn TE, Atkinson JB, Stewart JR,
PubMed PMID: 22226492. Hammon Jr. JW, et al. Unique antithymocyte serum versus OKT3
[55] Russo MJ, Iribarne A, Hong KN, Ramlawi B, Chen JM, for induction immunotherapy after heart transplantation. J Heart
Takayama H, et al. Factors associated with primary graft failure Transplant 1990;9(5):489 94; PubMed PMID: 2121921.
after heart transplantation. Transplantation 2010;90(4):444 50; [69] Kirklin JK, Bourge RC, White-Williams C, Naftel DC, Thomas
PubMed PMID: 20622755. FT, Thomas JM, et al. Prophylactic therapy for rejection after car-
[56] Stehlik J, Edwards LB, Kucheryavaya AY, Aurora P, Christie JD, diac transplantation. A comparison of rabbit antithymocyte globu-
Kirk R, et al. The Registry of the International Society for Heart lin and OKT3. J Thorac Cardiovasc Surg 1990;99(4):716 24;
and Lung Transplantation: 27th official adult heart transplant PubMed PMID: 2108282.
report—2010. J Heart Lung Transplant 2010;29(10):1089 103; [70] Macdonald PS, Mundy J, Keogh AM, Chang VP, Spratt PM. A
PubMed PMID: 20870164. prospective randomized study of prophylactic OKT3 versus
[57] Marasco SF, Esmore DS, Negri J, Rowland M, Newcomb A, equine antithymocyte globulin after heart transplantation—
Rosenfeldt FL, et al. Early institution of mechanical support increased morbidity with OKT3. Transplantation 1993;55
improves outcomes in primary cardiac allograft failure. J Heart (1):110 6; PubMed PMID: 8380508.
Lung Transplant 2005;24(12):2037 42; PubMed PMID: [71] Mehra MR, Zucker MJ, Wagoner L, Michler R, Boehmer J,
16364846. Kovarik J, et al. A multicenter, prospective, randomized, double-
[58] Listijono DR, Watson A, Pye R, Keogh AM, Kotlyar E, Spratt P, blind trial of basiliximab in heart transplantation. J Heart Lung
et al. Usefulness of extracorporeal membrane oxygenation for Transplant 2005;24(9):1297 304; PubMed PMID: 16143248.
422 PART | IV Heart

[72] Segovia J, Rodriguez-Lambert JL, Crespo-Leiro MG, Almenar L, [84] Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacody-
Roig E, Gomez-Sanchez MA, et al. A randomized multicenter namics of mycophenolate in solid organ transplant recipients. Clin
comparison of basiliximab and muromonab (OKT3) in heart trans- Pharmacokinet 2007;46(1):13 58; PubMed PMID: 17201457.
plantation: SIMCOR study. Transplantation 2006;81(11):1542 8; [85] Kobashigawa J, Miller L, Renlund D, Mentzer R, Alderman E,
PubMed PMID: 16770243. Bourge R, et al. A randomized active-controlled trial of mycophe-
[73] Mattei MF, Redonnet M, Gandjbakhch I, Bandini AM, Billes A, nolate mofetil in heart transplant recipients. Mycophenolate mofe-
Epailly E, et al. Lower risk of infectious deaths in cardiac trans- til investigators. Transplantation 1998;66(4):507 15; PubMed
plant patients receiving basiliximab versus anti-thymocyte globu- PMID: 9734496.
lin as induction therapy. J Heart Lung Transplant 2007;26 [86] Kaczmarek I, Ertl B, Schmauss D, Sadoni S, Knez A, Daebritz S,
(7):693 9; PubMed PMID: 17613399. et al. Preventing cardiac allograft vasculopathy: long-term benefi-
[74] Carrier M, Leblanc MH, Perrault LP, White M, Doyle D, cial effects of mycophenolate mofetil. J Heart Lung Transplant
Beaudoin D, et al. Basiliximab and rabbit anti-thymocyte globulin 2006;25(5):550 6; PubMed PMID: 16678034.
for prophylaxis of acute rejection after heart transplantation: a [87] Dandel M, Jasaityte R, Lehmkuhl H, Knosalla C, Hetzer R.
non-inferiority trial. J Heart Lung Transplant 2007;26(3):258 63; Maintenance immunosuppression with mycophenolate mofetil:
PubMed PMID: 17346628. long-term efficacy and safety after heart transplantation.
[75] Hershberger RE, Starling RC, Eisen HJ, Bergh CH, Kormos RL, Transplant Proc 2009;41(6):2585 8; PubMed PMID:
Love RB, et al. Daclizumab to prevent rejection after cardiac 19715979.
transplantation. N Engl J Med 2005;352(26):2705 13; PubMed [88] Keogh A, Richardson M, Ruygrok P, Spratt P, Galbraith A,
PMID: 15987919. O’Driscoll G, et al. Sirolimus in de novo heart transplant recipi-
[76] Teuteberg JJ, Shullo M, Zomak R, McNamara D, McCurry K, ents reduces acute rejection and prevents coronary artery disease
Kormos RL. Aggressive steroid weaning after cardiac transplanta- at 2 years: a randomized clinical trial. Circulation 2004;110
tion is possible without the additional risk of significant rejection. (17):2694 700; PubMed PMID: 15262845.
Clin Transplant 2008;22(6):730 7; PubMed PMID: 18673374. [89] Eisen HJ, Tuzcu EM, Dorent R, Kobashigawa J, Mancini D,
[77] Singh TP, Faber C, Blume ED, Worley S, Almond CS, Smoot Valantine-von Kaeppler HA, et al. Everolimus for the prevention
LB, et al. Safety and early outcomes using a corticosteroid- of allograft rejection and vasculopathy in cardiac-transplant reci-
avoidance immunosuppression protocol in pediatric heart trans- pients. N Engl J Med 2003;349(9):847 58; PubMed PMID:
plant recipients. J Heart Lung Transplant 2010;29(5):517 22; 12944570.
PubMed PMID: 20061164. [90] Kertesz NJ, Towbin JA, Clunie S, Fenrich AL, Friedman RA,
[78] Taylor DO, Barr ML, Radovancevic B, Renlund DG, Mentzer Jr. Kearney DL, et al. Long-term follow-up of arrhythmias in pediat-
RM, Smart FW, et al. A randomized, multicenter comparison of ric orthotopic heart transplant recipients: incidence and correlation
tacrolimus and cyclosporine immunosuppressive regimens in car- with rejection. J Heart Lung Transplant 2003;22(8):889 93;
diac transplantation: decreased hyperlipidemia and hypertension PubMed PMID: 12909469.
with tacrolimus. J Heart Lung Transplant 1999;18(4):336 45; [91] Drury JH, Labovitz AJ, Miller LW. Echocardiographic guidance
PubMed PMID: 10226898. for endomyocardial biopsy. Echocardiography 1997;14
[79] Reichart B, Meiser B, Vigano M, Rinaldi M, Martinelli L, (5):469 74; PubMed PMID: 11174985.
Yacoub M, et al. European multicenter tacrolimus (FK506) heart [92] Baraldi-Junkins C, Levin HR, Kasper EK, Rayburn BK,
pilot study: one-year results—European tacrolimus multicenter Herskowitz A, Baughman KL. Complications of endomyocardial
heart study group. J Heart Lung Transplant 1998;17(8):775 81; biopsy in heart transplant patients. J Heart Lung Transplant
PubMed PMID: 9730426. 1993;12(Pt 1):63 7; PubMed PMID: 8443204.
[80] Grimm M, Rinaldi M, Yonan NA, Arpesella G, Arizon Del Prado [93] Saraiva F, Matos V, Goncalves L, Antunes M, Providencia LA.
JM, Pulpon LA, et al. Superior prevention of acute rejection by Complications of endomyocardial biopsy in heart transplant
tacrolimus vs. cyclosporine in heart transplant recipients—a large patients: a retrospective study of 2117 consecutive procedures.
European trial. Am J Transplant 2006;6(6):1387 97; PubMed Transplant Proc 2011;43(5):1908 12; PubMed PMID: 21693299.
PMID: 16686762. [94] Fiorelli AI, Coelho GH, Aiello VD, Benvenuti LA, Palazzo JF,
[81] Ye F, Ying-Bin X, Yu-Guo W, Hetzer R. Tacrolimus versus Santos Junior VP, et al. Tricuspid valve injury after heart transplan-
cyclosporine microemulsion for heart transplant recipients: a tation due to endomyocardial biopsy: an analysis of 3550 biopsies.
meta-analysis. J Heart Lung Transplant 2009;28(1):58 66; Transplant Proc 2012;44(8):2479 82; PubMed PMID: 23026624.
PubMed PMID: 19134532. [95] Pham MX, Teuteberg JJ, Kfoury AG, Starling RC, Deng MC,
[82] Baran DA, Zucker MJ, Arroyo LH, Alwarshetty MM, Ramirez Cappola TP, et al. Gene-expression profiling for rejection surveil-
MR, Prendergast TW, et al. Randomized trial of tacrolimus mono- lance after cardiac transplantation. N Engl J Med 2010;362
therapy: tacrolimus in combination, tacrolimus alone compared (20):1890 900; PubMed PMID: 20413602.
(the TICTAC trial). J Heart Lung Transplant 2007;26(10):992 7; [96] Mehra MR, Kobashigawa JA, Deng MC, Fang KC, Klingler TM,
PubMed PMID: 17919618. Lal PG, et al. Clinical implications and longitudinal alteration of
[83] Kobashigawa JA, Miller LW, Russell SD, Ewald GA, Zucker MJ, peripheral blood transcriptional signals indicative of future cardiac
Goldberg LR, et al. Tacrolimus with mycophenolate mofetil allograft rejection. J Heart Lung Transplant 2008;27(3):297 301;
(MMF) or sirolimus vs. cyclosporine with MMF in cardiac trans- PubMed PMID: 18342752.
plant patients: 1-year report. Am J Transplant 2006;6 [97] Starling RC, Pham M, Valantine H, Miller L, Eisen H, Rodriguez
(6):1377 86; PubMed PMID: 16686761. ER, et al. Molecular testing in the management of cardiac
Chapter | 28 Current Status of Heart Transplantation 423

transplant recipients: initial clinical experience. J Heart Lung 4000 implants and counting. J Heart Lung Transplant 2012;31
Transplant 2006;25(12):1389 95; PubMed PMID: 17178330. (2):117 26; PubMed PMID: 22305376.
[98] Kobashigawa JA. The future of heart transplantation. Am J [110] Miller LW, Pagani FD, Russell SD, John R, Boyle AJ, Aaronson
Transplant 2012;12(11):2875 91; PubMed PMID: 22900830. KD, et al. Use of a continuous-flow device in patients awaiting
[99] Mehra MR, Parameshwar J. Gene expression profiling and car- heart transplantation. N Engl J Med 2007;357(9):885 96;
diac allograft rejection monitoring: is IMAGE just a mirage? J PubMed PMID: 17761592.
Heart Lung Transplant 2010;29(6):599 602; PubMed PMID: [111] Copeland JG, Smith RG, Arabia FA, Nolan PE, Sethi GK, Tsau
20497885. PH, et al. Cardiac replacement with a total artificial heart as a
[100] Kobashigawa JA, Kiyosaki KK, Patel JK, Kittleson MM, Kubak bridge to transplantation. N Engl J Med 2004;351(9):859 67;
BM, Davis SN, et al. Benefit of immune monitoring in heart PubMed PMID: 15329423.
transplant patients using ATP production in activated lympho- [112] Copeland JG, Copeland H, Gustafson M, Mineburg N,
cytes. J Heart Lung Transplant 2010;29(5):504 8; PubMed Covington D, Smith RG, et al. Experience with more than 100
PMID: 20133166. total artificial heart implants. J Thorac Cardiovasc Surg
[101] Kobashigawa JA, Katznelson S, Laks H, Johnson JA, Yeatman 2012;143(3):727 34; PubMed PMID: 22245242.
L, Wang XM, et al. Effect of pravastatin on outcomes after car- [113] Suarez J, Patel CB, Felker GM, Becker R, Hernandez AF,
diac transplantation. N Engl J Med 1995;333(10):621 7; Rogers JG. Mechanisms of bleeding and approach to patients
PubMed PMID: 7637722. with axial-flow left ventricular assist devices. Circ Heart Fail
[102] Wenke K, Meiser B, Thiery J, Nagel D, von Scheidt W, 2011;4(6):779 84; PubMed PMID: 22086831.
Steinbeck G, et al. Simvastatin reduces graft vessel disease and [114] Uriel N, Pak SW, Jorde UP, Jude B, Susen S, Vincentelli A,
mortality after heart transplantation: a four-year randomized et al. Acquired von Willebrand syndrome after continuous-
trial. Circulation 1997;96(5):1398 402; PubMed PMID: flow mechanical device support contributes to a high preva-
9315523. lence of bleeding during long-term support and at the time of
[103] Wenke K, Meiser B, Thiery J, Nagel D, von Scheidt W, Krobot transplantation. J Am Coll Cardiol 2010;56(15):1207 13;
K, et al. Simvastatin initiated early after heart transplantation: 8- PubMed PMID: 20598466.
year prospective experience. Circulation 2003;107(1):93 7; [115] John R, Lietz K, Schuster M, Naka Y, Rao V, Mancini DM,
PubMed PMID: 12515749. et al. Immunologic sensitization in recipients of left ventricular
[104] Schroeder JS, Gao SZ. Calcium blockers and atherosclerosis: les- assist devices. J Thorac Cardiovasc Surg 2003;125(3):578 91;
sons from the Stanford transplant coronary artery disease/diltia- PubMed PMID: 12658200.
zem trial. Can J Cardiol 1995;11(8):710 5; PubMed PMID: [116] Patlolla V, Patten RD, Denofrio D, Konstam MA, Krishnamani
7671182. R. The effect of ventricular assist devices on post-transplant
[105] Gupta A, Mancini D, Kirtane AJ, Kaple RK, Apfelbaum MA, mortality an analysis of the United network for organ sharing
Kodali SK, et al. Value of drug-eluting stents in cardiac trans- thoracic registry. J Am Coll Cardiol 2009;53(3):264 71;
plant recipients. Am J Cardiol 2009;103(5):659 62; PubMed PubMed PMID: 19147043.
PMID: 19231329. [117] John R, Pagani FD, Naka Y, Boyle A, Conte JV, Russell SD, et
[106] Bhama JK, Nguyen DQ, Scolieri S, Teuteberg JJ, Toyoda Y, al. Post-cardiac transplant survival after support with a
Kormos RL, et al. Surgical revascularization for cardiac allograft continuous-flow left ventricular assist device: impact of dura-
vasculopathy: Is it still an option? J Thorac Cardiovasc Surg tion of left ventricular assist device support and other variables.
2009;137(6):1488 92; PubMed PMID: 19464469. J Thorac Cardiovasc Surg 2010;140(1):174 81; PubMed
[107] DeBakey ME. Left ventricular bypass pump for cardiac assis- PMID: 20447659.
tance. Clinical experience. Am J Cardiol 1971;27(1):3 11; [118] Kirklin JK, Naftel DC, Kormos RL, Stevenson LW, Pagani FD,
PubMed PMID: 5538711. Miller MA, et al. Second INTERMACS annual report: more
[108] Kirklin JK, Naftel DC, Kormos RL, Stevenson LW, Pagani FD, than 1000 primary left ventricular assist device implants. J Heart
Miller MA, et al. Fifth INTERMACS annual report: Risk factor Lung Transplant 2010;29(1):1 10; PubMed PMID: 20123242.
analysis from more than 6000 mechanical circulatory support [119] Shah MR, Starling RC, Schwartz Longacre L, Mehra MR,
patients. J Heart Lung Transplant 2013;32(2):141 56; PubMed Working Group. Heart transplantation research in the next
PMID: 23352390. decade—a goal to achieving evidence-based outcomes: National
[109] Kirklin JK, Naftel DC, Kormos RL, Stevenson LW, Pagani FD, Heart, Lung, and Blood Institute Working Group. J Am Coll
Miller MA, et al. The Fourth INTERMACS Annual Report: Cardiol 2012;59:1263 9.

You might also like