evolving areas in heart...

10
Evolving Areas in Heart Transplantation Brittany N. Weber, MD, a Jon A. Kobashigawa, MD, b Michael M. Givertz, MD a ABSTRACT It has been 50 years since Dr. Christiaan Barnard performed the rst human-to-human heart transplant in December 1967 in South Africa. Remarkable progress has been made since that time, including changes in surgical techniques, immunosuppression, donor and recipient selection, and post-transplant care. In this paper, we provide a perspective on the changing face of heart transplantation and highlight key evolving areas. Topics that are covered include advances in immunosuppression, screening for acute and chronic rejection, cardiac allograft vasculopathy, and ongoing advancements in cardiac replacement therapy, including xenotransplantation, stem-cell research, tissue engineering, and the total articial heart. (J Am Coll Cardiol HF 2017;5:86978) © 2017 by the American College of Cardiology Foundation. T his month marks the 50th anniversary since the rst human-to-human heart transplant was performed by Dr. Christiaan Barnard in South Africa in December 1967. Although a ground- breaking moment in history for cardiovascular medi- cine, this did not come as a surprise to the medical and research community, as the groundwork had been laid since the early 1900s. In celebration of this anniversary, we offer the second part of a 2-part series that discusses advances in immunosuppression and evolving areas of complementary and alternative therapies in the future of cardiac transplantation (Central Illustration). NOVEL IMMUNOSUPPRESSION In transplant biology, immunosuppression is primarily targeted against the adaptive immune system, which involves both T and B cells. Many novel potential therapeutic targets exist (Table 1), but have not been prospectively studied in the cardiac transplant population. Belatacept is a fusion protein composed of the Fc fragment of a human immunoglobulin (Ig) G1 linked to the extracellular domain of CTLA-4, which blocks T-cell costimula- tion. It has been studied in a randomized controlled trial (RCT) in renal transplant recipients in 2 different doses compared with cyclosporine (CSA). At 12 months, survival was similar between the groups. The belatacept groups had higher glomerular ltration rates, but more episodes of rejection, post- transplant lymphoproliferative disorder, and tuber- culosis infection. Belatacept is currently approved for calcineurin inhibitor (CNI)-free regimens in the renal population (1). The Janus kinasesignal transducer and activator of transcription signaling pathway is important for immune activation. Tofacitinib is the rst inhibitor of Janus kinase that is currently approved for treat- ment of rheumatoid arthritis. It has been compared with tacrolimus in the renal transplant population and found to be equally effective (2). Compared with CSA, tofacitinib was associated with improved renal function, but also with a higher risk of infection and post-transplant lymphoproliferative disorder (3). Sotrastaurin is an agent that blocks early T-cell activation through inhibition of protein kinase C. It has been studied in both preclinical (rat) cardiac transplant models and renal transplant patients and was found to prolong graft survival. However, higher rates of rejection in a CNI-free regimen were observed (4). Tocilizumab (Actemra, Genentech, South San Francisco, California) is a humanized monoclonal antibody against the interleukin-6 receptor that is currently approved for refractory inammatory dis- eases. In preclinical animal models, it reduces allograft rejection, expands T-regulatory cell population, and reduces memory B-cell response. In highly-sensitized renal transplant recipients, tocilizumab reduces From the a Cardiovascular Division, Brigham and Womens Hospital, Harvard Medical School, Boston, Massachusetts; and the b Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California. Dr. Kobashigawa has received research grants and/or research support from Novartis, CareDx, Alexion, and TransMedics. Drs. Weber and Givertz have reported that they have no relationships relevant to the contents of this paper to disclose. Manuscript received August 24, 2017; revised manuscript received October 3, 2017, accepted October 10, 2017. JACC: HEART FAILURE VOL. 5, NO. 12, 2017 ª 2017 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION PUBLISHED BY ELSEVIER ISSN 2213-1779/$36.00 https://doi.org/10.1016/j.jchf.2017.10.009

Upload: others

Post on 31-Dec-2019

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7

ª 2 0 1 7 B Y T H E AM E R I C A N C O L L E G E O F C A R D I O L O G Y F O U N D A T I O N

P U B L I S H E D B Y E L S E V I E R

I S S N 2 2 1 3 - 1 7 7 9 / $ 3 6 . 0 0

h t t p s : / / d o i . o r g / 1 0 . 1 0 1 6 / j . j c h f . 2 0 1 7 . 1 0 . 0 0 9

Evolving Areas in Heart Transplantation

Brittany N. Weber, MD,a Jon A. Kobashigawa, MD,b Michael M. Givertz, MDa

ABSTRACT

FrobC

an

no

Ma

It has been 50 years since Dr. Christiaan Barnard performed the first human-to-human heart transplant in December

1967 in South Africa. Remarkable progress has been made since that time, including changes in surgical techniques,

immunosuppression, donor and recipient selection, and post-transplant care. In this paper, we provide a perspective on

the changing face of heart transplantation and highlight key evolving areas. Topics that are covered include advances in

immunosuppression, screening for acute and chronic rejection, cardiac allograft vasculopathy, and ongoing advancements in

cardiac replacement therapy, including xenotransplantation, stem-cell research, tissue engineering, and the total artificial

heart. (J Am Coll Cardiol HF 2017;5:869–78) © 2017 by the American College of Cardiology Foundation.

T his month marks the 50th anniversary sincethe first human-to-human heart transplantwas performed by Dr. Christiaan Barnard in

South Africa in December 1967. Although a ground-breaking moment in history for cardiovascular medi-cine, this did not come as a surprise to the medicaland research community, as the groundwork hadbeen laid since the early 1900s. In celebration of thisanniversary, we offer the second part of a 2-partseries that discusses advances in immunosuppressionand evolving areas of complementary and alternativetherapies in the future of cardiac transplantation(Central Illustration).

NOVEL IMMUNOSUPPRESSION

In transplant biology, immunosuppression isprimarily targeted against the adaptive immunesystem, which involves both T and B cells. Manynovel potential therapeutic targets exist (Table 1),but have not been prospectively studied in thecardiac transplant population. Belatacept is a fusionprotein composed of the Fc fragment of a humanimmunoglobulin (Ig) G1 linked to the extracellulardomain of CTLA-4, which blocks T-cell costimula-tion. It has been studied in a randomized controlledtrial (RCT) in renal transplant recipients in 2different doses compared with cyclosporine (CSA).At 12 months, survival was similar between thegroups. The belatacept groups had higher glomerular

m the aCardiovascular Division, Brigham and Women’s Hospital, Harva

edars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, Calif

d/or research support from Novartis, CareDx, Alexion, and TransMedics. D

relationships relevant to the contents of this paper to disclose.

nuscript received August 24, 2017; revised manuscript received October 3

filtration rates, but more episodes of rejection, post-transplant lymphoproliferative disorder, and tuber-culosis infection. Belatacept is currently approvedfor calcineurin inhibitor (CNI)-free regimens in therenal population (1).

The Janus kinase–signal transducer and activatorof transcription signaling pathway is important forimmune activation. Tofacitinib is the first inhibitorof Janus kinase that is currently approved for treat-ment of rheumatoid arthritis. It has been comparedwith tacrolimus in the renal transplant populationand found to be equally effective (2). Compared withCSA, tofacitinib was associated with improved renalfunction, but also with a higher risk of infection andpost-transplant lymphoproliferative disorder (3).Sotrastaurin is an agent that blocks early T-cellactivation through inhibition of protein kinase C. Ithas been studied in both preclinical (rat) cardiactransplant models and renal transplant patients andwas found to prolong graft survival. However, higherrates of rejection in a CNI-free regimen wereobserved (4).

Tocilizumab (Actemra, Genentech, South SanFrancisco, California) is a humanized monoclonalantibody against the interleukin-6 receptor that iscurrently approved for refractory inflammatory dis-eases. In preclinical animalmodels, it reduces allograftrejection, expands T-regulatory cell population, andreduces memory B-cell response. In highly-sensitizedrenal transplant recipients, tocilizumab reduces

rd Medical School, Boston, Massachusetts; and the

ornia. Dr. Kobashigawa has received research grants

rs. Weber and Givertz have reported that they have

, 2017, accepted October 10, 2017.

Page 2: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

ABBR EV I A T I ON S

AND ACRONYMS

ACR = acute cellular rejection

AMR = antibody-mediated

rejection

CMR = cardiac magnetic

resonance

CNI = calcineurin inhibitor

CSA = cyclosporine

EMB = endomyocardial biopsy

IgG = immunoglobulin G

LGE = late gadolinium

enhancement

Weber et al. J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7

Evolving Areas in Heart Transplantation D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8

870

alloantibody levels (5). Eculizumab, a hu-manized immunoglobulin G2/4 monoclonalantibody that inhibits complement by bindingto complement component C5, could also bebeneficial in antibody-mediated rejection(AMR). It reduced the incidence and severityof AMR in renal and lung transplant recipients(6,7); however, eculizumab has not beenstudied in heart transplantation, and cost willlikely be a barrier to widespread use.

Last, an enzyme from streptococcus pyo-genes, IdeS, is a unique agent that cleaveshuman IgG antibodies. In highly-sensitizedrenal transplant patients, it has been

shown to be effective in reducing alloantibody levels(8), with 2 further ongoing phase 2 clinical trials(NCT02790437 and NCT02426684). These agents allhave the potential to be utilized in cardiac transplantrecipients, and the field awaits studies in this popu-lation and potential future application.

SCREENING FOR ACUTE AND

CHRONIC REJECTION

Early detection of cardiac allograft rejection iscrucial for post-transplant care. Despite progress inimmunosuppression, acute cellular rejection (ACR)and AMR remain serious complications during andafter the first post-transplant year. Patients withearly treated rejection are at higher risk of latemorbidity and mortality (9). The current gold stan-dard for diagnosis is endomyocardial biopsy (EMB),although this is not ideal due to invasive risk, sam-pling error, and inter-reader variability. Despitedecades of basic and clinical research, biomarkersand imaging have been limited in their ability todiagnose and monitor treatment of rejection.Therefore, there is currently an unmet need tocreate an objective diagnostic test for cardiac allo-graft rejection.

NONINVASIVE IMAGING. Transthoracic echocardi-ography is a useful tool to monitor hemodynamically-significant rejection given wide availability and lowcost. Disadvantages include operator dependence andinter-reader variability. Many studies have examinedsystolic and diastolic predictive indexes of ACR (10).Although no single parameter can reliably diagnoserejection, if 1 or more parameters is abnormal, theprobability of ACR is higher. Three-dimensionalechocardiography can provide more accurate andreproducible measures of ventricular size and func-tion, but is not widely available. Global longitudinalstrain has been shown to be a predictor of subclinicalrejection (11). Studies of tissue Doppler parameters,

such as peak systolic and diastolic wall motion ve-locity, have yielded mixed results in predicting ACR.

Quantitative contrast myocardial perfusion echo-cardiography is a technique that can provide bothstructural and functional parameters and assess themicrovasculature. In a murine model of heart trans-plantation, contrast-enhanced echocardiographydemonstrated a decline in microvascular perfusion inmice that developed acute or chronic rejection(Figure 1). Notably, myocardial perfusion was restoredfollowing immunosuppressive therapy (12). A proof-of-concept study is currently underway to assess theefficacy of contrast-enhanced ultrasonography indetecting heart transplant rejection in humans(NCT02300870).

Nuclear imaging techniques have been studiedas a means for noninvasive screening of acute rejec-tion. 18FFluorodeoxyglucose positron emission to-mography imaging has been demonstrated to detectallograft rejection in a murine transplant model (13).Challenges in human transplantation include therequired protocol to suppress normal myocyteglucose consumption by a high-fat carbohydrate diet,and confounding diagnoses such as infectionand malignancy. Many other agents that targetcellular-specific components have been examinedin preclinical models, including 111indium-labeledantibody to myosin, 99mtechnetium-labeled annexin-V, and 111indium labeled oligonucleotides thatrecognize interleukin-2—all markers that are overex-pressed in acute rejection (14,15). None of theseagents, however, has been evaluated in large-scalehuman studies, and thus, the clinical translation isnot yet known.

Cardiac magnetic resonance (CMR) imaging is apromising screening modality for detection of rejec-tion. CMR lacks ionizing radiation and can survey theentire myocardium, decreasing the possibility offalse negatives due to sampling error as observed inEMB. T2-weighted imaging has been most widelyused, as T2 values prolong with higher water content,which increases in edema and inflammation (16). In astudy evaluating the diagnostic accuracy of CMRversus EMB for acute rejection, CMR had a highsensitivity (93%) and high negative predictivevalue (98%), highlighting that CMR could be utilizedas a screening test before routine EMB (17). Thepresence of late gadolinium enhancement (LGE) hasalso been investigated in small cohorts (18), but therole in transplant screening is not clear in theabsence of prospective data with larger samplesizes from multiple centers. Investigational contrastagents are also being developed to improve thesensitivity for detection of inflammation. Ultrasmall

Page 3: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

CENTRAL ILLUSTRATION Evolving Areas of Heart Transplantation

Weber, B.N. et al. J Am Coll Cardiol HF. 2017;5(12):869–78.

This central illustration demonstrates 3 broad areas discussed in this review: novel immunosuppressive agents to treat allograft rejection,

new modalities in imaging and genomic medicine to screen for rejection, and future potential ways to increase the “donor” pool.

J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7 Weber et al.D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8 Evolving Areas in Heart Transplantation

871

superparamagnetic iron oxide (USPIO) particles canbe injected intravenously, and following ingestionby macrophages, detected on T2-weighted CMR.This promising technique has been studied in ratmodels, but thus far, no human translational studiesexist (19).

A novel test that measures markers of oxidativestress has also been developed to assist in detection ofcardiac allograft rejection. Allograft rejection results in

increased production of myocardial reactive oxygenspecies, which are then degraded by lipid peroxidationand secreted as volatile organic compounds. TheHeartsbreath test (Menssana Research, Newark, NewJersey) analyzes the methylated alkane contour inexpired air that has been identified as a marker of sig-nificant (grade 3R) rejection (20). A prospectivemulticenter clinical trial is currently underway tovalidate these findings (NCT01397812).

Page 4: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

TABLE 1 Novel Therapeutic Targets for Immunosuppression

Target Agent(s) Efficacy Risks/Barriers

Blockade of T-cell costimulationvia CTLA-4

Belatacept Similar survival compared to CSA in renal transplant patientsLess renal toxicity

Higher rates of rejection, PTLD, and TB infection

JAK-STAT inhibition Tofacitinib Effective in renal transplant patients compared with TAC and CSALess renal toxicity

Higher rates of infection and PTLD

Inhibition of protein kinase C Sotrastaurin Prolonged graft survival in pre-clinical cardiac studies andrenal transplant recipients

Higher rates of rejection compared to CNI regimenDose-dependent gastrointestinal toxicity

Inhibition of complement bybinding to C5

Eculizumab Reduction in AMR in renal and lung transplant patients Substantial cost

IL-6 receptor antagonist Tocilizumab Reduced allograft rejection and expansion of regulatoryT cells in animal models

Reduced alloantibody levels in renal transplant patients

No clinical studies in cardiac transplant recipients

Cleavage of human IgG antibodies IdeS Reduced alloantibody levels in sensitized renaltransplant patients

No clinical studies in cardiac transplant recipients

AMR ¼ antibody-mediated rejection; CNI ¼ calcineurin inhibitor; CSA ¼ cyclosporine; IgG ¼ immunoglobulin G; JAK-STAT ¼ Janus kinase–signal transducer and activator of transcription; IL ¼ interleukin;PTLD ¼ post-transplant lymphoproliferative disorder; TAC ¼ tacrolimus; TB ¼ tuberculosis.

Weber et al. J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7

Evolving Areas in Heart Transplantation D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8

872

GENOMIC MEDICINE AND IMMUNE ASSAYS. A newfrontier in cardiac transplant screening is the promiseof genomic medicine. Approaches currently underinvestigation include donor-derived cell-free deoxy-ribonucleic acid (DNA), microRNA, and gene expres-sion profiling. The AlloMap (CareDx, Brisbane,California) is a gene expression profile of peripheral

FIGURE 1 Use of MP Mapping to Detect Acute Cardiac Allograft Rej

Parametric perfusion maps of syngeneic and allogeneic cardiac allograft

groups compared with syngeneic groups over time. On day 8 post-trans

allogeneic hearts. Red and yellow represent higher echo-intensity chang

from John Wiley & Sons, Inc.

blood that has been incorporated into the Interna-tional Society of Heart and Lung Transplantation(ISHLT) guidelines (21). The test screens for a 20-genepanel with 11 rejection-related genes and 9 genes fornormalization and quality control. The result is re-ported as a single score on a scale of 0 to 40, whichindicates the probability of moderate or severe ACR

ection

s show a progressive decline in tissue perfusion in the allogeneic

plant, microvascular perfusion (MP) was markedly reduced in

e and higher MP. Reprinted from Fischer et al. (12) with permission

Page 5: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

TABLE 2 Current Scorecard on Xenotransplantation

Advantages Disadvantages

Unlimited organ supply Size mismatch

Elective availability Ethical restrictions on primate use

Expansion of candidate pool Ecological restrictions on species use

Avoid effects of brain death oncardiac function

Hyperacute, acute, and chronic vascularrejection

Potential for genetic manipulation Coagulation dysfunction

Cost saving Transmission of zoonoses

J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7 Weber et al.D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8 Evolving Areas in Heart Transplantation

873

based on findings from the CARGO (Cardiac AllograftRejection Gene Expression Observational) study. The11 genes were identified based on the highest andmost statistically significant expression differences(22). The landmark IMAGE (Invasive MonitoringAttenuation through Gene Expression) trial randomlyassigned 602 low-risk patients to either AlloMap orEMB for routine rejection surveillance. Rates of allo-graft dysfunction, death, or retransplant were similarbetween groups, and patient satisfaction was higherin the AlloMap group due to less biopsies (23). How-ever, AlloMap is only predictive for ACR and notAMR, and has not proven effective for early screeningor in patients who are at higher risk for late rejection.

Graft-derived cell-free deoxyribonucleic acid(GcfDNA) has emerged as another possible biomarker.After transplantation and organ engraftment, GcfDNAwill reach its highest value up to >5% of total cfDNA,with a rapid decrease to <0.5% within 1 week. Duringan episode of rejection, GcfDNA are shed into the bloodand can be accompanied by a 5-fold increase that canbe measured by current assays. The release of GcfDNAoccurs earlier than other clinical markers of rejectionand could potentially be used to monitor transplantpatients at regular intervals to allow early diagnosisand treatment (24). Because GcfDNA and AlloMapmeasure different signals in the blood, a combinationof the 2 approaches could be additive, as AlloMap is ameasure of host immune response and GcfDNA moni-tors graft injury. Pilot studies are underway to comparethese 2 approaches to rejection monitoring.CARDIAC ALLOGRAFT VASCULOPATHY. Cardiacallograft vasculopathy (CAV), also known as chronicrejection, remains the leading cause of late mortalityfollowing heart transplantation resulting in 1 of 8deaths beyond 1 year (9). A complex interplayof immune-mediated inflammation and endothelialinjury and dysfunction ultimately leads to intimalhyperplasia andmicrovascular/epicardial disease (25).

Routine surveillance is critical for diagnosis ofCAV, as many patients are asymptomatic. Invasivecoronary angiography remains the standard of careand is a Class I screening recommendation; however,there is robust interest in newer noninvasive imagingtechniques for diagnosis and management. ISHLTgrading by coronary angiography is categorized into 3stages defined by severity of left main, primary, andbranch vessel stenoses and presence or absence ofallograft dysfunction (LVEF <45%) and restrictivephysiology (26). Noninvasive imaging has also beenutilized, but has not replaced invasive angiographydue to lower diagnostic accuracy. Currently availabletests (followed by ISHLT recommendation) includedobutamine stress echocardiography (Class IIa),

single-photon emission computed tomography (CT)(Class IIa), and coronary CT angiography (Class IIb).A normal dobutamine stress echocardiography hasa moderate negative predictive value (80% to 100%)for subsequent cardiac events (27). Similarly, single-photon emission CT imaging has moderate diag-nostic accuracy (sensitivity 63% to 84%, specificity70% to 78%) to detect minor to severe stenoses thatmay be secondary to a limited ability to detectbalanced ischemia, which often underlies CAV pa-thology (28). More recently, myocardial flow reserveassessed by 82rubidium positron emission tomogra-phy imaging has been shown to predict clinical out-comes post-heart transplant, although multicenterdata with long-term follow-up is lacking (29).

Dobutamine is the most frequently used pharma-cological stressor, with a sensitivity of 70% to 80% todetect significant CAV compared with coronary angi-ography. Exercise is less predictive due to chronotropicincompetence fromcardiac denervation,with a limitedsensitivity of 15% to 33%. Last, Doppler echocardiog-raphy can be used to calculate coronary flow reserve(CFR), which correlates with invasively-measuredcoronary flow reserve and has been shown to havehigh sensitivity and specificity for detecting CAV (11).

FUTURE PROSPECTS AND

ACTIVE AREAS OF RESEARCH

XENOTRANSPLANTATION. Xenotransplantation re-fers to the process of grafting or transplanting organsor tissues between members of different species.With increasing numbers of patients with end-stageheart failure and shortage of donors, there has beena renewed interest in the possibility of cardiac xeno-transplantation (Table 2) (30). Advantages include anunlimited supply and elective availability of donororgans, expansion of the candidate pool, and avoid-ance of detrimental effects of brain death on donororgan function. Although monkeys and baboons aremost phylogenetically like humans, given theecological and ethical restrictions, the pig is the onlyanimal available to replace human tissues. The pig is

Page 6: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

FIGURE 2 Cardiopoietic Cell Therapy for Advanced Ischemic Heart Failure

(A) The primary efficacy outcome in CHART-1 (see text). The Mann-Whitney estimator, or

probability that the treatment group had a better outcome on the composite primary

endpoint, was 0.54 (95% confidence interval [CI]: 0.47 to 0.61; p ¼ 0.27). The

corresponding Mann-Whitney odds was 1.17 (95% CI: 0.89 to 1.55). (B) The primary

efficacy outcome in the subgroup of patients with left ventricular end-diastolic volume

(LVEDV) 200 to 370ml. The Mann-Whitney estimator, or probability that the treatment

group had a better outcome on the composite primary endpoint, was 0.61 (95% CI: 0.52

to 0.70; p ¼ 0.015). Corresponding Mann-Whitney odds was 1.57 (95% CI: 1.09 to 2.35).

Reprinted from Bartunek et al. (39) with permission from Oxford University Press on

behalf of the European Society of Cardiology.

Weber et al. J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7

Evolving Areas in Heart Transplantation D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8

874

physiologically similar to humans and organ size iscomparable; it can be raised in controlled environ-ments and, more importantly, genetically modified.Main limitations to xenotransplantation are rejection,including hyperacute, acute, and chronic vascular,and risk of transmitting zoonoses. The developmentof genetically-modified pigs has allowed for theexpression of human antigens that can reduce the riskof rejection and provides hope to future feasibility.

Hyperacute rejection is a result of antibodiesagainst anti-aGal composed of polyclonal glycopro-teins that are naturally present in humans andrepresent 1% of circulating IgG. Porcine endothelialcells express Gal-a1, 3-Gal, which results in activationof the complement cascade and coagulation system.In turn, endothelial edema, interstitial hemorrhage,vascular deposit of immunoglobulins, and microvas-cular thrombosis occur within minutes to hours.Various techniques to counteract this response,including antibody removal through plasmapheresisor inhibition of complement cascade, have beendescribed. Although hyperacute rejection can beavoided, acute humoral rejection still occurs. Bothbarriers have been addressed with genetically engi-neered pigs. Pigs have been created with deletion ofthe enzyme (a1,3-galactosyltransferase) that pro-duces Gal (GTKO) and are transgenic for 1 or morehuman complement regulatory proteins (CRPs) (31).These proteins, including CD46, CD55, and CD59 (30),prevent the development of an acute humoral

response, whereas deletion of Gal blocks the hyper-acute response. Despite success with this approach,the T-cell mediated response also needs to be tar-geted to prevent graft failure. Strategiesinclude conventional immunosuppression and“costimulation” blockade. Genetic engineering with amutant major histocompatibility complex II or bytransgenic expression of an immunosuppressanttarget into transgenic pigs has demonstrated somesuccess (32).

However, even if the hyperacute, acute humoral,and T-cell responses are successfully blocked, graftsurvival remains limited to weeks or months sec-ondary to coagulation dysfunction, with featuressimilar to thrombotic microangiopathy. A similartheme of utilizing the power of genetic engineeringhas focused on the introduction of human anticoag-ulant and antithrombotic agents into GTKO/CRP pigs.Encouraging early results have been published in 2baboons with transplantation of GTKO/CD46/throm-bomodulin pig heart grafts with no features of graftvasculopathy after 1 to 2 years (33).

Studies discussed above are inOldWorld nonhumanprimates. Nonhuman primates express the oligosac-charide N-glycolylneuraminic acid (NeuGc) thatresults in antipig antibodies in humans. Before trans-lation to humans, pigs will need to be developed thatlack NeuGc. Fortunately, these studies are currentlyunderway (30,34).

STEM CELL THERAPY AND TISSUE ENGINEERING. Cellular-based therapy and tissue engineering are promisingapproaches to reversal of advanced heart failure.Landmark scientific work has demonstrated thatresident cardiomyocytes have the ability for self-renewal; annual cardiomyocyte turnover rate is w1%in young adults and decreases to 0.5% in the elderly(35). Promoting this endogenous repair process hasthe potential to enhance regenerative capacity andmodify or reverse adverse LV remodeling. Regenera-tive cell-based therapies ultimately aim to restorenormal myocardial function through both directcell-mediated and indirect paracrine-mediated repairmechanisms.

Initial enthusiasm, however, has been dampenedby inconsistent and modest benefits in clinical trials.These studies, which have been performed in acutemyocardial infarction, ischemic and dilated cardio-myopathy, and advanced heart failure, have beenextensively summarized (36). Differences in trialdesign, source of cardiac stem cell or progenitor cells,cell number and technique, and small sample sizesare some of the factors that have led to discrepantresults. However, recent meta-analyses, which now

Page 7: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

FIGURE 3 Functional Maturation of EHM

(A) Force-frequency response of engineered human

myocardium (EHM) (at 4 weeks in culture) generated by the

starting protocol (red) that included a variety of undefined

matrix (Matrigel) and serum (horse serum, fetal calf serum,

chick embryo extract) components (n ¼ 8), and the serum-

free protocol (black) containing 4% B27 without insulin plus

transferring growth factor-b1, insulin growth factor-1,

fibroblast growth factor-2, and vascular endothelial growth

factor (n ¼ 21). *p < 0.05 by 2-way repeated measures

analysis of variance (ANOVA) followed by Sidak multiple

comparison test; §p < 0.05 vs. 1 Hz of the respective group

by 2-way ANOVA with the Tukey multiple comparison post

hoc test. (B) Representative force traces recorded from

EHM (at 4 weeks in culture) at 1.5-Hz stimulation with an

intermittent stimulation pause (10 s); enhanced force of

contraction (FOC) at the reintroduction of electric

stimulation (i.e., post-rest potentiation) is characteristic

for cardiomyocytes with mature intracellular calcium

storage and release (dotted line indicates pre-pause

baseline maximal FOC). Reprinted from Tiburcy et al. (43),

with permission from Wolters Kluwer Health, Inc.

J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7 Weber et al.D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8 Evolving Areas in Heart Transplantation

875

include a larger number of better-designed studies,have hinted at an overall benefit. Fisher et al. (37)analyzed 31 RCTs, including a total of 1,521 patients,that used a range of cell types, including bonemarrow-derived mononuclear cells and mesenchymalstem cells, skeletal myoblasts, and adipose tissue-derived cells. In addition to safety, a reduction inmortality and rehospitalization during long-termfollow-up was observed along with modest improve-ments in ejection fraction and heart failure symp-toms. However, only one-half of the trials wereblinded, and one-half failed to report the method ofallocation concealment. When analyzed separately,double-blind studies showed no significant differ-ences, raising concern for bias skewing overallresults.

The isolation of refined cell types continues toevolve. Initial trials utilized “first-generation” cell-based therapies, which predominantly consisted ofunselected progenitor cells, mesenchymal stem cells,and endothelial progenitor cells from bone marrow.Although these populations have the potential todifferentiate into cell types within the myocardium,homing and differentiation are not guaranteed. Inresponse, the field has moved to “second- or next-generation” therapies. These include isolation andexpansion of cardiac resident stem cells or thecreation of cardiopoietic stem cells through lineage-specific differentiation of bone marrow–derivedprogenitors, commonly referred to as cardiopoiesis.Cardiopoietic stem cell therapies have demonstratedpromise in proof-of-concept studies, including theC-CURE (Cardiopoietic Stem Cell Therapy in HeartFailure) trial (38). However, a large pivotal trial,CHART-1 (Congestive Heart Failure CardiopoieticRegenerative Therapy), failed to confirm theseresults.

In the CHART-1 trial, 271 patients with ischemiccardiomyopathy were randomized to endocardialdelivery of cardiopoietic cells or sham procedure at 30centers. At 39 weeks, there was no significant differ-ence between groups in the primary hierarchicalcomposite endpoint of all-cause mortality, worseningheart failure, quality-of-life score, 6-min walk dis-tance, LV end-systolic volume, and ejection fraction(39). A subgroup analysis, however, did suggest abeneficial effect in patients with severe LV dilation(Figure 2) (40). Similar promising results were seen inthe ixCELL-DCM (Ixmyelocel-T Administered ViaTransendocardial Catheter-based Injections to Sub-jects With Heart Failure Due to Ischemic DilatedCardiomyopathy) study using a proprietary technique(Vericel, Cambridge, Massachusetts) that selectivelyexpands 2 types of bone marrow mononuclear cells

(CD90þ mesenchymal stem cells and CD45þ CD14þ

autofluorescentþ activated macrophages). In thisdouble-blind RCT, 109 patients with advancedischemic cardiomyopathy were randomized to

Page 8: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

FIGURE 4 Total Cardiac Replacement for Irreversible Biventricular Heart Failure

(A) Carmat total artificial heart (TAH). Advantages include a blood-contacting side of membrane comprised of pericardial bovine tissue,

bioprosthetic valves, and sensors to measure flow and pressure; disadvantages include its complex design, risk of valve or chamber failure,

several moving parts, bulky size, and high risk of thromboembolism. (B) BiVACOR TAH. Advantages include magnetic bearings, compact size

with single rotational component, high-flow capabilities, controller to balance left and right blood flows, and low thrombus risk; disad-

vantages include the complex control system to maintain centered location of rotating impeller. Adapted from Fox et al. (46) with

permission from John Wiley & Sons, Inc.

Weber et al. J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7

Evolving Areas in Heart Transplantation D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8

876

receive cell therapy or placebo via transendocardialdelivery. At 12 months, there was a 37% reduction inthe primary endpoint of death, cardiovascular hos-pitalization, or clinic visits for worsening heart failurein the cell therapy group (p ¼ 0.034), and there weremore serious adverse events in the placebo group(p ¼ 0.02) (41).

Cardiac tissue engineering is another novelapproach to regenerative therapy that uses engi-neered human myocardium (EHM) (42). The seminalfindings that human embryonic stem cells andhuman-induced pluripotent stem cells can bedirected into organ-specific cell types, along withavailability and scalability, have led to their use asbona fide cardiomyocytes in tissue engineering. Thechallenge of EHM is to create functional car-diomyocytes with the electromechanical properties ofhuman myocardium. In a recent study, cellular andchemical components for EHM generation wereoptimized, resulting in advanced cardiomyocytematuration (43). Also, for the first time, a positiveforce frequency and other evidence of functionalmaturation were demonstrated (Figure 3). In-vestigators also transferred EHM into nude rats usingan epicardial approach leading to revascularization ofthe grafts. However, electromechanical coupling tonormal myocardium was not clearly seen. Although

not yet ready for clinical trials, EHM is a promisingapproach to cardiac repair that may soon be feasible.

The future success of regenerative medicine willdepend on scientists and clinicians working togetherto achieve validity and utility with the long-term goalof widespread use. Quality control manufacturingand delivery of products in a uniform way are criticalto this effort. In addition, patient-specific factors thatcan affect regenerative fitness, such as age, sex, andcomorbidities, must be accounted for. Not every in-dividual harbors stem cells with uniform regenerativecapacity, and tools are needed to predict the likeli-hood of effectiveness at the patient level.TOTAL ARTIFICIAL HEART. The development of atotal artificial heart (TAH) to replace the failing hearthas been a focus of global research for decades. Thefirst successful use of a TAH was by Dr. Denton Cooleyin 1969 in a patient bridged to heart transplant.Although the patient died within 36 h of transplantsurgery, proof of concept was demonstrated. Sincethen, multiple devices have been developed withlimited success. The only device currently approvedby the U.S. Food and Drug Administration is the70cc SynCardia Temporary Total Artificial Heart(TAH-t) (SynCardia Systems, Tucson, Arizona). TheTAH-t is currently indicated as a bridge to transplantin patients with irreversible biventricular failure,

Page 9: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7 Weber et al.D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8 Evolving Areas in Heart Transplantation

877

refractory hemodynamic instability, or ventriculararrhythmias, and a contraindication to left ventricu-lar assist device. Despite the marked increase in leftventricular assist device use, demand for the TAHremains low. According to the Interagency Registryfor Mechanically Assisted Circulatory Support, therewere 22 TAH implants in 2007, and this increased toonly 54 in 2014 (44). The TAH-t consists of 2pneumatically-operated chambers that provide totalsystemic and pulmonary flow. A large pneumaticdriver supplies pulses of compressed air to the leftand right chambers, with unidirectional flow (up to 10l/min) achieved with 4 titling valves. Patients requireanticoagulation and are at risk of thrombotic orhemorrhagic stroke. Infections are common due totube exit sites as portals of entry. Despite the devel-opment of a portable driver, published experiencewith discharge to home is limited to only 11 patientswith 88 readmissions (81% unplanned) after a medianfollow-up of 349 days (45).

In 2013, the U.S. Food and Drug Administrationapproved 2 humanitarian use device designations fora smaller 50cc TAH-t to be used for destination ther-apy and pediatric bridge-to-transplant. Thus far, 28devices have been implanted and there is currently anongoing 3-arm clinical trial to investigate safety andbenefit to both pediatric and adult transplant-eligiblepatients (NCT02459054). Patients with congenitalheart disease and advanced heart failure may beespecially appropriate for this smaller device. Otherdevices currently under development for total cardiacreplacement include the Carmat TAH (Carmat, Vilizy-Villacoublay, France) (Figure 4A), a continuous-flow

TAH being developed at the Cleveland Clinic with aunique design that is part bovine and part machine;and the BiVACOR device (BiVACOR, Houston, Texas)(Figure 4B), which uses a third-generation magneticsuspension technology (46).

CONCLUSIONS

It has been 50 years since Dr. Christiaan Barnardperformed the first human-to-human heart transplantin South Africa. Advances in immunosuppression,donor and recipient selection, surgical techniques,and post-transplant care have resulted in excellentmedium- and long-term outcomes for the majority ofpatients with end-stage heart failure currently un-dergoing transplant. However, significant barriersremain, including early and late risks of organ rejec-tion and vasculopathy along with continuous risk ofinfection and malignancy. Although the donor pool iscautiously being expanded, it is unlikely to ever fillthe needs of a continuously growing and sick patientpopulation. The future depends on further progressin stem cell research, tissue engineering, and xeno-transplantation, while optimizing post-transplanthealth through improved immunosuppression andprevention of acute and chronic rejection. Thereversal of heart failure to avoid the need for organreplacement is also paramount.

ADDRESS FOR CORRESPONDENCE: Dr. Michael M.Givertz, Cardiovascular Division, Brigham andWomen’s Hospital, 75 Francis Street, Boston,Massachusetts02115. E-mail:[email protected].

RE F E RENCE S

1. Durrbach A, Pestana JM, Pearson T, et al.A phase III study of belatacept versus cyclosporinein kidney transplants from extended criteriadonors (BENEFIT-EXT study). Am J Transplant2010;10:547–57.

2. Busque S, Leventhal J, Brennan DC, et al.Calcineurin-inhibitor-free immunosuppressionbased on the JAK inhibitor CP-690,550: a pilotstudy in de novo kidney allograft recipients. Am JTransplant 2009;9:1936–45.

3. Vincenti F, Silva HT, Busque S, et al. Evaluationof the effect of tofacitinib exposure on outcomesin kidney transplant patients. Am J Transplant2015;15:1644–53.

4. Russ GR, Tedesco-Silva H, Kuypers DR, et al.Efficacy of sotrastaurin plus tacrolimus after denovo kidney transplantation: randomized, phase IItrial results. Am J Transplant 2013;13:1746–56.

5. Vo AA, Choi J, Kim I, et al. A phase I/II trial of theinterleukin-6 receptor-specific humanized mono-clonal (tocilizumab)þ intravenous immunoglobulin

in difficult to desensitize patients. Transplantation2015;99:2356–63.

6. Dawson KL, Parulekar A, Seethamraju H.Treatment of hyperacute antibody-mediated lungallograft rejection with eculizumab. J Heart LungTransplant 2012;31:1325–6.

7. Stegall MD, Diwan T, Raghavaiah S, et al. Ter-minal complement inhibition decreases antibody-mediated rejection in sensitized renal transplantrecipients. Am J Transplant 2011;11:2405–13.

8. Jordan SC, Lorant T, Choi J, et al. IgG endo-peptidase in highly sensitized patients undergoingtransplantation. N Engl J Med 2017;377:442–53.

9. Lund LH, Edwards LB, Dipchand AI, et al. TheRegistry of the International Society for Heart andLung Transplantation: Thirty-third Adult HeartTransplantation Report-2016; Focus Theme: Pri-mary Diagnostic Indications for Transplant. J HeartLung Transplant 2016;35:1158–69.

10. Dandel M, Hetzer R. Post-transplant surveil-lance for acute rejection and allograft

vasculopathy by echocardiography: usefulness ofmyocardial velocity and deformation imaging.J Heart Lung Transplant 2017;36:117–31.

11. Badano LP, Miglioranza MH, Edvardsen T, et al.European Association of Cardiovascular Imaging/Cardiovascular Imaging Department of the Brazil-ian Society of Cardiology recommendations for theuse of cardiac imaging to assess and follow pa-tients after heart transplantation. Eur Heart JCardiovasc Imaging 2015;16:919–48.

12. Fischer K, Ohori S, Meral FC, et al. Testing the ef-ficacy of contrast-enhanced ultrasound in detectingtransplant rejection using a murine model of hearttransplantation. Am J Transplant 2017;17:1791–801.

13. Daly KP, Dearling JLJ, Seto T, et al. Use of[18F]FDG PET to monitor the development ofcardiac allograft rejection. Transplantation 2015;99:e132–9.

14. Gierthmuehlen M, Sommer P, Zuhayra M, et al.Imaging of acute heart-transplant rejection using99m-Technetium labelled oligonucleotides

Page 10: Evolving Areas in Heart Transplantationheartfailure.onlinejacc.org/content/jhf/5/12/869.full.pdfEvolving Areas in Heart Transplantation Brittany N. Weber, MD, aJon A. Kobashigawa,

Weber et al. J A C C : H E A R T F A I L U R E V O L . 5 , N O . 1 2 , 2 0 1 7

Evolving Areas in Heart Transplantation D E C E M B E R 2 0 1 7 : 8 6 9 – 7 8

878

against interleukin-2 mRNA in rats. Eur J Car-diothorac Surg 2010;37:1111–6.

15. Narula J, Acio ER, Narula N, et al. Annexin-Vimaging for noninvasive detection of cardiac allo-graft rejection. Nat Med 2001;7:1347–52.

16. Usman AA, Taimen K, Wasielewski M, et al.Cardiac magnetic resonance T2 mapping in themonitoring and follow-up of acute cardiac trans-plant rejection: a pilot study. Circ Cardiovasc Im-aging 2012;5:782–90.

17. Butler CR, Savu A, Bakal JA, et al. Correlation ofcardiovascular magnetic resonance imaging findingsand endomyocardial biopsy results in patients un-dergoing screening for heart transplant rejection.J Heart Lung Transplant 2015;34:643–50.

18. Taylor AJ, Vaddadi G, Pfluger H, et al. Diag-nostic performance of multisequential cardiacmagnetic resonance imaging in acute cardiacallograft rejection. Eur J Heart Fail 2010;12:45–51.

19. Wu YL, Ye Q, Sato K, Foley LM, Hitchens TK,Ho C. Noninvasive evaluation of cardiac allograftrejection by cellular and functional cardiac magneticresonance. J Am Coll Cardiol Img 2009;2:731–41.

20. Phillips M, Boehmer JP, Cataneo RN, et al.Heart allograft rejection: detection with breathalkanes in low levels (the HARDBALL study).J Heart Lung Transplant 2004;23:701–8.

21. Costanzo MR, Dipchand A, Starling R, et al. TheInternational Society of Heart and Lung Trans-plantation guidelines for the care of heart transplantrecipients. JHeart LungTransplant 2010;29:914–56.

22. Deng MC, Eisen HJ, Mehra MR, et al. Nonin-vasive discrimination of rejection in cardiac allo-graft recipients using gene expression profiling.Am J Transplant 2006;6:150–60.

23. Pham MX, Teuteberg JJ, Kfoury AG, et al.Gene-expression profiling for rejection surveil-lance after cardiac transplantation. N Engl J Med2010;362:1890–900.

24. Beck J, Oellerich M, Schulz U, et al. Donor-derived cell-free DNA is a novel universalbiomarker for allograft rejection in solid organtransplantation. Transplant Proc 2015;47:2400–3.

25. Chih S, Chong AY, Mielniczuk LM, Bhatt DL,Beanlands RSB. Allograft vasculopathy: theAchilles’ heel of heart transplantation. J Am CollCardiol 2016;68:80–91.

26. Mehra MR, Crespo-Leiro MG, Dipchand A,et al. International Society for Heart and LungTransplantation working formulation of a

standardized nomenclature for cardiac allograftvasculopathy–2010. J Heart Lung Transplant2010;29:717–27.

27. Spes CH, Klauss V, Mudra H, et al. Diag-nostic and prognostic value of serial dobut-amine stress echocardiography for noninvasiveassessment of cardiac allograft vasculopathy: acomparison with coronary angiography andintravascular ultrasound. Circulation 1999;100:509–15.

28. Manrique A, Bernard M, Hitzel A, et al.Diagnostic and prognostic value of myocardialperfusion gated SPECT in orthotopic hearttransplant recipients. J Nucl Cardiol 2010;17:197–206.

29. Mc Ardle BA, Davies RA, Chen L, et al. Prog-nostic value of rubidium-82 positron emission to-mography in patients after heart transplant. CircCardiovasc Imaging 2014;7:930–7.

30. Murthy R, Bajona P, Bhama JK, Cooper DKC.Heart xenotransplantation: historical background,experimental progress, and clinical prospects. AnnThorac Surg 2016;101:1605–13.

31. Phelps CJ, Koike C, Vaught TD, et al. Produc-tion of alpha 1,3-galactosyltransferase-deficientpigs. Science 2003;299:411–4.

32. Hara H, Witt W, Crossley T, et al. Humandominant-negative class II transactivator trans-genic pigs—effect on the human anti-pig T-cellimmune response and immune status. Immu-nology 2013;140:39–46.

33. Mohiuddin MM, Singh AK, Corcoran PC, et al.One-year heterotopic cardiac xenograft survivalin a pig to baboon model. Am J Transplant 2014;14:488–9.

34. Lutz AJ, Li P, Estrada JL, et al. Doubleknockout pigs deficient in N-glycolylneuraminicacid and galactose a-1,3-galactose reduce thehumoral barrier to xenotransplantation. Xeno-transplantation 2013;20:27–35.

35. Bergmann O, Bhardwaj RD, Bernard S, et al.Evidence for cardiomyocyte renewal in humans.Science 2009;324:98–102.

36. Behfar A, Crespo-Diaz R, Terzic A, Gersh BJ.Cell therapy for cardiac repair—lessons from clin-ical trials. Nat Rev Cardiol 2014;11:232–46.

37. Fisher SA, Doree C, Mathur A, Martin-Rendon E. Meta-analysis of cell therapy trials forpatients with heart failure. Circ Res 2015;116:1361–77.

38. Bartunek J, Behfar A, Dolatabadi D, et al.Cardiopoietic stem cell therapy in heart failure: theC-CURE (Cardiopoietic stem Cell therapy in heartfailURE) multicenter randomized trial withlineage-specified biologics. J Am Coll Cardiol2013;61:2329–38.

39. Bartunek J, Terzic A, Davison BA, et al.Cardiopoietic cell therapy for advanced ischaemicheart failure: results at 39weeksof the prospective,randomized, double blind, sham-controlledCHART-1 clinical trial. Eur Heart J 2017;38:648–60.

40. Teerlink JR, Metra M, Filippatos GS, et al.Benefit of cardiopoietic mesenchymal stemcell therapy on left ventricular remodelling:results from the Congestive Heart FailureCardiopoietic Regenerative Therapy (CHART-1)study. Eur J Heart Fail 2017 May 31 [E-pubahead of print].

41. Patel AN, Henry TD, Quyyumi AA, et al.Ixmyelocel-T for patients with ischaemic heartfailure: a prospective randomised double-blindtrial. Lancet 2016;387:2412–21.

42. Yanamandala M, Zhu W, Garry DJ, et al.Overcoming the roadblocks to cardiac cell therapyusing tissue engineering. J Am Coll Cardiol 2017;70:766–75.

43. Tiburcy M, Hudson JE, Balfanz P, et al. Definedengineered human myocardium with advancedmaturation for applications in heart failuremodeling and repair. Circulation 2017;135:1832–47.

44. Kirklin JK, Naftel DC, Pagani FD, et al. SeventhINTERMACS annual report: 15,000 patients andcounting. J Heart Lung Transplant 2015;34:1495–504.

45. Quader MA, Green AJ, Shah KB, Cooke R,Kasirajan V. Hospital readmissions after dischargeto home with the Total Artificial Heart Freedomdriver: readmission reasons, clinical outcomes, andhealth care costs. J Heart Lung Transplant 2016;35:251–2.

46. Fox CS, McKenna KL, Allaire PE,Mentzer RM, Throckmorton AL. Total artificialhearts—past, current, and future. J Card Surg2015;30:856–64.

KEY WORDS cardiac allograftvasculopathy, heart transplantation,immunosuppression, stem cells, tissueengineering, transplant rejection