ArticlePDF Available

Abstract and Figures

Introduction: Improved survival following single ventricle palliation has led to a large population of patients with a Fontan circulation, many of whom will eventually develop Fontan failure. Many of these patients will require heart transplantation. However, more than half of these patients have Fontan failure despite preserved ventricular function. Increasing experience with ventricular assist devices (VAD) in children has paved the way for VAD support in those with failing univentricular circulation. Areas covered: The use of VADs to support the failing univentricular circulation is a relatively new concept. Most studies have focussed on supporting patients with the failing systemic ventricle. However, there are limited reports on VAD support of the pulmonary circulation in those patients with a failing Fontan circulation despite preserved ventricular function. Although the concept of implantation of a "mechanical right ventricle" is attractive, none of the current VADs have been designed to support the low-pressure pulmonary circulation. Novel devices, which use low-pressure, high-flow pumps, specifically designed to support the pulmonary circulation, are currently under development. However, at this stage none have been tested in vivo. Expert commentary: The failing univentricular circulation is one of the great challenges in the field of congenital heart disease. While current VADs are designed to support systemic circulation, many patients may require support of the pulmonary circulation in addition to systemic circulatory support or in isolation. A fully implantable VAD for support of the pulmonary circulation as destination therapy would be beneficial for patients with preserved systolic function, but must have a low energy requirement, negligible risk of stroke and low risk of device thrombosis and failure.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=ierd20
Download by: [The University Of Melbourne Libraries] Date: 06 November 2017, At: 17:37
Expert Review of Medical Devices
ISSN: 1743-4440 (Print) 1745-2422 (Online) Journal homepage: http://www.tandfonline.com/loi/ierd20
Ventricular assist devices for the failing
univentricular circulation
Edward Buratto, William Y. Shi, Xin Tao Ye & Igor E. Konstantinov
To cite this article: Edward Buratto, William Y. Shi, Xin Tao Ye & Igor E. Konstantinov (2017)
Ventricular assist devices for the failing univentricular circulation, Expert Review of Medical
Devices, 14:6, 449-459, DOI: 10.1080/17434440.2017.1332523
To link to this article: http://dx.doi.org/10.1080/17434440.2017.1332523
Accepted author version posted online: 22
May 2017.
Published online: 26 May 2017.
Submit your article to this journal
Article views: 57
View related articles
View Crossmark data
REVIEW
Ventricular assist devices for the failing univentricular circulation
Edward Buratto, William Y. Shi, Xin Tao Ye and Igor E. Konstantinov
Department of Cardiac Surgery, The Royal Childrens Hospital, Melbourne, Australia; Department of Paediatrics, The University of Melbourne,
Melbourne, Australia; Murdoch Childrens Research Institute, Melbourne, Australia
ABSTRACT
Introduction: Improved survival following single ventricle palliation has led to a large population of
patients with a univentricular circulation, many of whom develop heart failure. Increasing experience
with ventricular assist devices (VAD) in children has paved the way for VAD support in those with failing
univentricular circulation.
Areas covered: The use of VADs to support the failing univentricular circulation is a relatively new
concept. Most studies have focused on supporting patients with the failing systemic ventricle. There are
limited reports of VAD support of the pulmonary circulation in patients with Fontan failure despite
preserved ventricular function. None of the current VADs have been designed to support the pulmon-
ary circulation. Novel low-pressure, high-flow pumps, specifically designed to support the pulmonary
circulation, are under development.
Expert commentary: The failing univentricular circulation is one of the great challenges in the field of
congenital heart disease. While current VADs are designed to support the systemic circulation, many
patients require support of the pulmonary circulation. A fully implantable VAD for support of the
pulmonary circulation as destination therapy would be beneficial for patients with preserved systolic
function, but must have low energy requirements, negligible risk of stroke and low risk of device
thrombosis and failure.
ARTICLE HISTORY
Received 1 March 2017
Accepted 16 May 2017
KEYWORDS
Univentricular circulation;
Fontan failure; ventricular
assist devices; Fontan
circulation; single ventricle
palliation
1. Introduction
1.1. The univentricular circulation
The current standard of treatment for patients with a func-
tional single ventricle is multistage palliation, resulting in a
Fontan circulation [1,2]. The absence of a subpulmonary ven-
tricle results in elevated central venous pressure (CVP) that, in
turn, contributes to failure of the univentricular circulation
and, in particular, Fontan failure, despite preserved ventricular
function [13]. While improvements in surgical technique and
postoperative care have meant that early survival has
improved in recent years [48], it appears inevitable that
many univentricular circulations will eventually fail [13].
Heart transplantation in patients with a Fontan circulation is
technically challenging [9,10], but recent reports have demon-
strated improving results, comparable to other forms of con-
genital heart disease [1113]. Nevertheless, there is limited
donor supply and substantial mortality while awaiting trans-
plantation [14]. Thus, ventricular assist devices (VADs) are an
emerging option for the management of the failing univen-
tricular circulation (Figure 1), both to reduce waiting list mor-
tality [1416] and, potentially, as a destination therapy [2]. We
review the current status of VAD therapy in patients with
univentricular circulation and discuss the principles guiding
the application VAD technology in these patients.
1.2. Ventricular dysfunction in the univentricular
circulation
Systemic ventricular dysfunction may cause failure of the uni-
ventricular circulation at any stage of palliation. Ventricular fail-
ure in these patients tends to be multifactorial [17]. Firstly, the
single ventricle palliated with a systemic to pulmonary artery
shunt is subjected to excessive volume work and dilatation,
increasing stress on the growing ventricle [18]. The presence
of relatively poor cardiac output and low blood pressure con-
tributes to vasoconstriction, increased peripheral resistance and
hence increased afterload [18]. Secondly, multiple operations
with prolonged cross-clamp times cause ischemiareperfusion
injury to the myocardium, contributing to long-term ventricular
dysfunction [17]. In fact, prolonged cross-clamp time during the
Fontan procedure has been associated with risk of Fontan fail-
ure [19]. Thirdly, the systemic ventricle may be of right ventri-
cular morphology and hence not developmentally suited to
withstand systemic ventricular pressures [3,20]. Finally, in asso-
ciation with the patients structural heart disease, there may be
genetic mutations affecting the myocardium itself [17]. Hence,
ventricular dysfunction in patients with univentricular circula-
tion is multifactorial and challenging to manage. The use of
VADs to support the failing systemic ventricle has emerged as a
new treatment option for these patients.
CONTACT Igor E. Konstantinov igor.konstantinov@rch.org.au The Royal Childrens Hospital, Flemington Road, Parkville, Melbourne, VIC 3052, Australia
EXPERT REVIEW OF MEDICAL DEVICES, 2017
VOL. 14, NO. 6, 449459
https://doi.org/10.1080/17434440.2017.1332523
© 2017 Informa UK Limited, trading as Taylor & Francis Group
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
2. VADs in the univentricular circulation
2.1. Current status of VADs
In adults, VADs have an established role in the treatment of
end-stage heart failure as a bridge-to-transplantation, destina-
tion therapy, or rarely, recovery [21]. Over an 8-year period,
the INTERMACS registry has documented the implantation of
over 15,000 VADs in North America, of which 60.9% were
implanted as bridge-to-transplantation and 38.2% as destina-
tion therapy [21]. Over this short period, there has been an
expansion of indications, refinement in technology and
implantation strategies, improvement in survival and reduc-
tion in complications [2124]. One of the most important
advances has been the development of continuous flow
VADs, which are associated with lower rates of complications
and improved survival [21,25]. Recent experience from the
INTERMACS registry has demonstrated a 1-year survival of
80% for left ventricular assist devices (LVAD) and 50% for
biventricular assist devices (biVAD) [21].
In contrast, in the pediatric population, VAD therapy is
more challenging due to patient size as well as the complex
anatomy associated with congenital heart disease [26].
Nevertheless, there is evidence that the advent of VAD tech-
nology has reduced the risk of waiting list mortality in children
listed for heart transplantation [1416] and improved survival
compared to extracorporeal membrane oxygenation (ECMO)
[26]. The only available VAD developed specifically for children
is the Berlin Heart EXCOR (BHE), a pulsatile paracorporeal
device, which is suitable for supporting children with a body
weight as low as 2.5 kg [26,27]. Due to the improved out-
comes associated with the use of continuous flow devices in
adults, they have been increasingly used in children [28]. So
far the smallest child supported with a continuous flow VAD
(HeartWare) was 3.7 years of age, with a body surface area
(BSA) of 0.6 m
2
, weighing 13.5 kg [29]. The relationship
between patient size and selection of VAD technology is
demonstrated in Figure 2.
The first report of the PediMACS registry, a multicenter
database including pediatric VAD implantations performed at
66 North American centers, included 200 durable VADs, of
which just over half (109/200, 54.5%) were continuous flow
devices [31]. Almost all patients below 20 kg of weight were
supported by the BHE, while more than 80% (105/130) of
patients over 20 kg of weight were supported with continuous
flow devices [31]. At 6-month follow-up, 58% of patients had
been transplanted, 28% were alive on VAD support and 14%
had died. Survival of patients with continuous flow devices
was significantly better than those with pulsatile flow devices;
however, these patients were significantly older and had
higher BSA [31]. Nevertheless, the PediMACS registry demon-
strated that the rate of device malfunction was much higher
with the pulsatile flow devices [32], and as such continuous
flow VADs appear to be the device of choice for children large
enough to accommodate them.
Historically, Matusuda et al. [33] described the first
attempts at supporting children with univentricular circulation
with VADs in 1988. However, these initial attempts were
unsuccessful. The modern era of VAD support of the failing
univentricular circulation began in 2005, with Frazier et al. [34]
implanting a HeartMate VAD in a patient with Fontan failure.
To date, the total reported experience with VAD in patients
with univentricular circulation is 53 patients, which is summar-
ized in Table 1.
2.2. VAD support following stage I palliation
The experience of VAD support of patients following stage I
palliation is limited. The largest report is from Weinstein et al.
[50], who supported nine patients following stage I palliation,
of whom only one survived to transplantation. Specifically,
they had no survivors when VAD was implanted for failure to
wean from bypass in the operating theater or for failure to
wean from ECMO. The only survivor was 17-months post stage
I palliation at the time of VAD implantation. The authors
suggested that ECMO should be the preferred strategy in
this scenario due to the poor outcomes with VADs. Pearce
et al. [43] reported a 15-month-old boy with double outlet
right ventricle, d-transposition of the great arteries and mitral
atresia with circulatory failure following pulmonary artery
banding. The patient was treated with construction of a cen-
tral shunt, ligation of the pulmonary trunk and implantation of
BHE, with inlet cannula in the right atrium and outflow into
Figure 1. Algorithm for selection of the appropriate configuration of ventricular assist devices in the univentricular circulation.
450 E. BURATTO ET AL.
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
the ascending aorta. The patient was supported for 7 weeks
and then transplanted.
Support of children following stage I palliation has most
commonly been performed for postoperative ventricular dys-
function. There are numerous factors which may contribute to
the poor outcomes in this group, including the physiology of
the shunt-dependent circulation, patient age and size, and the
fact that support was generally used as salvage, due to inabil-
ity to wean from bypass. Given the poor results reported thus
far, VADs do not seem to provide an advantage over ECMO in
this setting.
2.3. VAD support following bidirectional cavopulmonary
shunt (BCPS)
There have been several reports of VAD support in patients
following BCPS, which are summarized in Table 1.The
largest experience is from Weinstein et al. [50], who
reported 12 patients supported with BHE VADs following
BCPS, of whom 58.3% (7/12) survived to transplantation.
Niebler et al. [52] described four patients treated with BHE
following BCPS, of whom 75% (3/4) survived to transplan-
tation. They found that despite achieving high cardiac out-
put, patients continued to have an elevated CVP, which
they attributed to abnormal aortopulmonary and veno-
venous connections. To overcome this challenge, they
advocated selecting a larger chamber size than would
normally be used for the same sized patient with biven-
tricular physiology [52]. Brancaccio et al. [47]reportedtwo
cases of VAD support following BCPS, of whom 50% (1/2)
survived to transplantation. Of a further four individual
cases described in the literature, 25% (1/4) survived to
transplantation. While the overall experience with VADs
in the setting of BCPS is limited, it appears to be a feasible
strategy with just over half of patients surviving to
transplantation.
2.4. Strategies of VAD support for the failing Fontan
Patients with Fontan failure fall into one of two categories:
those with impaired ventricular function and those with
preserved ventricular function [2]. The choice of VAD strat-
egy needs to be tailored to the nature of the patients
physiology. In initial reports of VAD therapy in the Fontan
circulation, the devices were used much like a conventional
LVAD, with the inflow cannula in the dominant ventricle or
atrium, and the outflow cannula in the ascending aorta. This
is the strategy of choice for patients with ventricular dys-
function as the cause of Fontan failure. It has been sug-
gested that this strategy may also work for patients with
predominantly right-sided failure in order to pullblood
flow across the pulmonary vascular bed [2]. However, it
may be ineffective in the setting of high venous pressures
due to increased pulmonary resistance, and so biventricular
support has been described for patients with both ventri-
cular dysfunction and elevated venous and pulmonary pres-
sures [35,49,51]. Furthermore, there is a group of Fontan
patients who primarily have high venous pressure and pul-
monary resistance as the cause of their failure, despite
preserved ventricular function. In this group, the use of
Figure 2. The relationship between choice of ventricular assist device, patient age and size. The use of continuous flow devices becomes increasingly prevalent as
age increases. Reproduced from Rossano J, Villa C, Konstantinov I [30], with permission.
EXPERT REVIEW OF MEDICAL DEVICES 451
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
Table 1. Reported experience of ventricular assist devices used to support failing univentricular circulation.
Author Year NDevice Age (years) Stage Preserved systolic function Duration of support (days) Outcome
Frazier [34] 2005 1 Heartmate IP LVAS 14 Fontan No 45 Transplantation
Nathan [35] 2006 1 BiVAD: Berlin Heart 4 Fontan Not reported 28 Transplantation
Newcomb [36] 2006 1 Thoratec 25 Fontan No 152 Transplantation
Calvaruso [37] 2007 1 Berlin Heart 10 Fontan No 7 Transplantation
Chu [38] 2007 1 Berlin Heart 4 BCPS No 10 Death
Pretre [39] 2008 1 RVAD Berlin Heart 27 Fontan Yes 395 Transplantation
Russo [40] 2008 1 Centrifugal pump 14 Fontan No >6 Transplantation
Cardarelli [41] 2009 1 Berlin Heart 1.5 Fontan No 183 Recovery
Irving [42] 2009 1 Berlin Heart 3 BCPS No 7 Transplantation
Pearce [43] 2009 1 Berlin Heart 1.3 PAB No 49 Transplantation
Meira [44] 2011 1 HeartWare 12 Fontan No 1 Transplantation
VanderPluym [45] 2011 1 Berlin Heart 3 Fontan Yes 174 Transplantation
Mackling [46] 2012 2 Berlin Heart 4 Fontan No 309 Death
4 BCPS No 270 Death
Brancaccio [47] 2013 2 Berlin Heart 2 BCPS No 2 Transplantation
Berlin Heart 4 BCPS No 166 Death
Sanders [48] 2014 1 Berlin Heart 16 Fontan No 2 Transplantation
Valeske [49] 2014 1 Berlin Heart BiVAD 19 Fontan No 23 Transplantation
Weinstein [50] 2014 26 Berlin Heart
LVAD = 24
BiVAD = 2
Stage 1 = 9 Not reported Median: Stage 1: transplantation in 1/9
Stage 2: transplantation in 7/12
Stage 3: transplantation in 3/5
Stage 2 = 12 52
Stage 3 = 5
Arnaoutakis [51] 2016 5 Heartware 18 Fontan Not reported Median: 60 (for survivors) Transplantation
TAH 14 Fontan Not reported Transplantation
Berlin heart 3 BCPS Not reported Death
Berlin Heart 5 Fontan Not reported Transplantation
Thoratec 23 Fontan Not reported Death
Niebler [52] 2016 4 Berlin Heart 0.6 2.3 BCPS Not reported 9 312 3 transplantations, 1 death
Total Experience 53 Stage 1 = 10 Stage 1: 2/10 (20%) transplanted
BCPS: 12/22 (55%) transplanted
Fontan: 16/21 (76%) transplanted
BCPS = 22
Fontan = 21
BCPS: bidirectional cavopulmonary shunt; LVAD: left ventricular assist device; BiVAD: biventricular assist device; PAB: pulmonary artery banding; RVAD: right ventricular assist device; TAH: total artificial heart.
452 E. BURATTO ET AL.
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
VADs to support the pulmonary circulation, so-called cavo-
pulmonary support, has been described [39]. These strate-
gies are demonstrated in Figures 3 and 4.
2.5. VADs to support the systemic ventricle
Inthemajorityofreportedcases, VADs have been used to support
the systemic ventricle in patients with univentricular physiology
[4548,5052], similarly to how an LVAD is used in the biventricu-
lar circulation (Figures 3(a) and 4(a)). In the majority of these cases,
BHE has been used (see Table 1), although there have also been
reports of Thoratec [36,51], HeartMate [34], and HeartWare [44].
The technique of VAD implantation to support the systemic ven-
tricle is similar to standard LVAD insertion; however, some specific
technical considerations have been described, particularly regard-
ing the positioning of the inlet cannula. Challenges include
adhesions due to previous surgery, coronary artery abnormalities,
presence of a prominent nondominant ventricle obscuring access
to the dominant ventricle, and cannula obstruction from ventri-
cular trabeculations and the subvalvular apparatus, especially
when the right ventricle is dominant [36,47,50,52]. Nevertheless,
in most reported cases, the inflow cannula has been placed in the
apex of the dominant left ventricle [36,37,41,42,48,5052]orinthe
diaphragmatic surface of the dominant right ventricle [38,44,47].
Additional strategies have been described to deal with inlet can-
nula obstruction, such as resection of muscular bands and resec-
tion of the atrioventricular valves and their subvalvular apparatus
[34,52]. Placement of the inflow cannula in the pulmonary venous
atrium has been described as another strategy to avoid inlet
cannula obstruction [33,40,51].
Outcomes of VAD support of the systemic ventricle have been
reported in two small case series and a number of case reports
Figure 3. Configurations of Berlin Heart Excor VAD in the Fontan circulation. (a) supporting the systemic ventricle, (b) supporting the pulmonary circulation and (c)
supporting both.
Figure 4. Configurations of continuous flow VAD in the Fontan circulation. (a) supporting the systemic ventricle, (b) supporting the pulmonary circulation and (c)
supporting both.
EXPERT REVIEW OF MEDICAL DEVICES 453
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
(Table 1). The largest report, from Weinstein et al. [50], includes
five patients with a Fontan circulation, of whom 60% (3/5) sur-
vived to transplantation. By comparison, 72% of children with
biventricular physiology from the same database survived to
transplantation. Arnatoukakis et al. [51]reportedfourpatients
with Fontan circulation who underwent VAD implantation, three
of whom (75%) survived to transplantation. Taken together, the
remaining case reports included 14 patients, of whom 78.6% (11/
14) survived to transplantation, 7.1% (1/14) were successfully
weaned and 14.3% died (2/14) [34,36,37,40,41,4446,48]. These
results are similar to those reported for the overall population of
children supported with VAD in the PediMACS registry, which
demonstrated that at 6-month follow-up, 61% of patients had
been transplanted, 31% remained on VAD support, and 8% had
died prior to transplant [32]. These results are encouraging and
seem to be similar to the overall results reported for VAD support
in the pediatric population. However, it must be recognized that
data from case reports are particularly prone to publication bias
toward good results, and that none of these reports contains
significant long-term follow-up.
The complications observed during VAD support were similar
to those seen with VAD in children with biventricular circula-
tions, dominated by bleeding, neurological events, and infection
[32]. Weinstein et al. [50] reported an overall adverse event rate
of 73.1%, with respiratory failure (42.3%), bleeding (38.5%),
infection (23.1%), and neurologic dysfunction (15.4%) being
the most common complications. Furthermore, pump change
due to thrombus was required in 26.9% of patients. For compar-
ison, in the PediMACS database, the commonest adverse events
were device malfunction (39.5%), infection (39.0%), bleeding
(34.0%), and neurological dysfunction (26%) [32].
Despite the technical challenge of implanting VADs in
patients with a univentricular circulation, survival to transplanta-
tion is in the range of 6080% across the small number of
published cases. Furthermore, the complication profile appears
to be similar to that reported for VADs in the general population
of children requiring ventricular support. The use of VAD ther-
apy as a bridge to transplantation for patients with a failing
univentricular circulation is emerging as a viable option.
2.6. Biventricularsupport in the univentricular
circulation
In patients with both ventricular dysfunction and raised venous
pressure and pulmonary resistance, supporting the systemic ven-
tricle may not be sufficient to achieve stable hemodynamics. In
these patients, it may be better to convert to biventricular sup-
port,with one pump supporting the systemic ventricle, and
another supporting the cavopulmonary circulation, much like a
conventional right ventricular assist device (RVAD) (Figures 3(c)
and 4(c)). There are few reports of biventricular support in the
setting of univentricular circulation, comprising three patients
with BiVADs and one patient with a total artificial heart (TAH).
Nathan et al. [35] reported BiVAD implantation in a 4-year-
old girl with a failing Fontan circulation. The child had plastic
bronchitis, pleural effusions, and a pulmonary vascular resis-
tance of 3.8 Wood units. Initially, a 30 mL BHE VAD was
implanted from the systemic ventricular apex to the ascending
aorta. However, as there was significant venous hypertension,
it was decided to place a 25 mL BHE as an RVAD. The cavo-
pulmonary anastomosis was taken down, the inflow cannula
was inserted into the lateral tunnel, and the outflow cannula
was inserted into the pulmonary arteries. This child underwent
transplantation after 28 days of support.
Valeske et al. [49] reported a 19-year-old male who had under-
gone Fontan with a total cavopulmonary connection (TCPC) and
presented in New York Heart Association (NYHA) class IV heart
failure with a severely dilated systemic ventricle. They implanted
two Berlin Heart pumps as a BiVAD. The RVAD drained both vena
cavae via the extracardiac conduit, with the outflow placed in the
pulmonary artery. The LVAD was placed between the left atrium
and the ascending aorta. The patient underwent successful
orthotopic heart transplantation after 23 days of support.
Arnaoutakis [51] reported a single case of TAH implantation
in a 14-year-old child with a failing Fontan circulation compli-
cated by renal failure and plastic bronchitis. They reported
recovery of end-organ function on TAH support and survival
to heart transplantation.
Although there is limited data on the use of BiVAD in
univentricular patients, there are promising results and this
strategy offers the potential to support patient with both a
failing systemic ventricle and high venous pressures, in whom
support of the systemic ventricle alone may not be sufficient.
2.7. Failure of Fontan circulation with preserved
ventricular function
Due to the lack of a sub-pulmonary ventricle, patients with a
Fontan circulation generally have a CVP of 1015 mmHg, three
times greater than the normalphysiologicallevel[1](Figure 5).
Hence, these patients are prone to complications of elevated
venous pressures, including hepatic congestion and cirrhosis,
ascites, peripheral edema, and protein-losing enteropathy [18].
Due to the low flow across the pulmonary vasculature, there is
chronic under-filling of the systemic ventricle, which in combina-
tion with ventricular hypertrophy, contributes to diastolic dysfunc-
tion [3,18]. Additionally, the systemic ventricle may also fail due to
the factors described earlier: dilatation and overloading prior to
BPCS, increased afterload, repeated cardiac procedures, genetic
mutations, and right ventricular morphology. All of these factors
may contribute to Fontan failure, which although lacking a stan-
dardized definition, is generally said to be any of death, transplan-
tation, takedown, Fontan conversion, NYHA class III/IV symptoms,
protein-losing enteropathy, or plastic bronchitis [53]. Based on the
mechanism of Fontan failure, patients may be classified into two
groups: those with ventricular dysfunction and those with pre-
served ventricular function [3]. Importantly, more than half of
patients with Fontan failure have preserved ejection fraction [20].
The importance of understanding the mode of Fontan failure is
highlighted by the fact that those with preserved ejection fraction
have poorer survival after transplantation [54], demonstrating that
patients will respond differently to interventions depending on
their underlying mechanism of Fontan failure.
2.8. Isolated support of pulmonary circulation
As has been previously described, more than half of patients
with a failing Fontan have preserved function of their
454 E. BURATTO ET AL.
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
dominant ventricle, and thus effectively have right-sidedfail-
ure [53]. These patients could be supported by a VAD placed
between the extracardiac conduit and the pulmonary arteries
(Figures 3(b) and 4(b)). There is only a single case report of
cavopulmonary support using a VAD in a patient with a uni-
ventricular circulation. Prêtre et al. [39] presented a 27-year-
old man who underwent Fontan conversion from an atriopul-
monary connection to extracardiac conduit and developed
severe cardiac failure 16 weeks later. The patient had normal
ventricular function but an elevated CVP of 33 mmHg. The
cavopulmonary anastomoses were taken down and the pul-
monary arteries repaired. The extracardiac conduit was
replaced with a larger graft and the superior vena cava (SVC)
and inferior vena cava (IVC) both anastomosed to it. The
inflow cannula of a 60 mL Berlin Heart was inserted into the
extracardiac conduit, while the outflow cannula was inserted
into the reconstructed pulmonary arteries. The patients clin-
ical condition improved substantially and he underwent car-
diac transplantation after 13 months of support. Given the
large proportion of Fontan patients with failure despite pre-
served ventricular function, this is a very promising strategy
and represents a proof of conceptfor the possibility of cavo-
pulmonary support for the treatment of Fontan failure.
However, the VADs currently available are designed for sup-
port of the systemic ventricle rather than the low-pressure
pulmonary circulation, and hence this strategy is currently
limited by the lack of appropriate pump technology.
3. Emerging technologies
While there have been encouraging results using VADs to support
the univentricular circulation, none of the devices currently avail-
able has been designed for this application. Given that more than
half of patients with Fontan failure effectively have right-sided
failure [53], with elevated venous pressures and pulmonary
resistance, much of the current research focuses on the develop-
ment of a cavopulmonary assist device. As previously discussed,
the major hemodynamic limitations of the Fontan circulation are
systemic venous hypertension, decreased pulmonary blood flow,
and under-filling of the systemic ventricle [55]. Based on the
hemodynamic complications observed with the Fontan circula-
tion, it has been postulated that the ideal hemodynamic effect of
the pump for cavopulmonary support is a small decrease in venous
pressure, with a small increase in pulmonary pressure and flow,
each of approximately 5 mmHg [5557]. As such, the ideal pump
for cavopulmonary support differs markedly from currently avail-
able VAD technology. Current VADs are designed to generate a
high-pressure gradient at partial flow rates, and as a result have
significant energy requirements and generate significant negative
pressures at their inflow cannula [2,55]. A device for cavopulmon-
ary support, which is a low-pressure, high-volume pump providing
a pressure step up of only 5 mmHg, is likely to be sufficient for
supporting the Fontan circulation [5557]. This should allow for
greater pump efficiency and improved battery life, potentially
allowing for a completely implantable design with a long-life
battery similar to pacemaker, or even transcutaneous charging
[2]. Finally, the ideal assist device would not obstruct the cavopul-
monary blood flow in the case of mechanical failure. Such devel-
opments would make destination therapy a viable option for
patients with Fontan failure.
Initial experimental research on cavopulmonary support of
the failing Fontan has utilized existing VAD technology in
animal models immediately after construction of the single-
stage TPCP. Several groups have shown that short-term sup-
port of the TCPC in an animal model is feasible, providing
adequate cardiac output and an increase in pulmonary arterial
(PA) pressures with stable or decreased venous pressures
compared to the biventricular circulation at baseline [55,57
62]. However, venous collapse and hence circulatory obstruc-
tion, as well as entrainment of air through the Goretex graft,
Figure 5. Diagrammatic representation of the haemodynamics of (a) the normal biventricular compared to (b) the Fontan circulation compared to. Note the
elevated central venous pressure due to the lack of a subpulmonary ventricle to drive pulmonary blood flow. Reprinted from Jaquiss and Aziz [2] with permission
from Elsevier.
EXPERT REVIEW OF MEDICAL DEVICES 455
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
have been observed due to the high negative pressures gen-
erated by these devices [58,59]. Furthermore, when placed in
the large caliber vessels of the TCPC, micro-axial pumps are
associated with recirculation of almost half of the pump flow,
as well as retrograde SVC flow, requiring higher pump speeds,
decreasing efficiency, and increasing the risk of hemolysis
[55,57,60].
To overcome the shortcomings of current generation VADs,
several groups are working on the development of novel
pumps specifically designed for cavopulmonary support.
Rodefelds group [63,64] have developed a design based on
the Von Kàrmàn impellar pump, which is implantable at the
junction of the TCPC, augmenting flow in all four directions,
directing outflow down both pulmonary arteries. Importantly
this is a low-power design, which provides a small step up in
pressure, replicating the role of the right ventricle.
Furthermore, even when stationary it improves efficiency of
the Fontan circulation by reducing turbulence, rather than
causing circulatory obstruction. This means that even in the
case of device failure it would still be of hemodynamic benefit,
which is an ideal situation. So far, however, only results of
computation modeling and in vitro experimentation have
been published.
Lacour-Gayet et al. [65] described the application of a novel
micro-axial flow pump in a modified in vitro mock-up of the
Fontan circulation. The extracardiac conduit in their model
had a Y-shaped configuration, with SVC and IVC inflow and
an outflow arm anastomosed to the pulmonary trunk. The
micro-axial pump was implanted in the outflow arm. Using
this strategy, recirculation and retrograde flow into the SVC
were minimized. In their model, CVP was reduced, PA pres-
sures were modestly increased, and cardiac output was
increased by up to 2L/min. However, they did observe collapse
of the vena cavae at pump speeds above 3000 rpm. This
design, however, has yet to be tested in vivo.
Throckmorton et al. [66] also described a novel, low-pres-
sure axial pump tested in vitro in a mock TCPC. The pump
demonstrated a pressure step up of 216 mmHg, without
evidence of cavitation. A further evolution of this pump is
designed to be percutaneously inserted in the IVC while pro-
viding the same hemodynamic benefits, and it has also been
tested in vitro [67]. Most recently, this group has published
work on refinements of axial flow pump design in order to
maximize efficiency for the specific application as cavopul-
monary support [68,69]. None of these designs has been
tested in vivo as of yet.
In vitro tests of existing VAD technology have provided
proof of concept for cavopulmonary support of the Fontan
circulation. However, these pumps have several limitations as
they are designed to support the systemic circulation. Novel
pumps designed specifically to address the requirement of
cavopulmonary support have shown promising results in
vitro, but have yet to be tested in vivo.
4. Conclusions
The failing Fontan circulation is one of the major challenges in
the field of congenital heart disease. There is a growing num-
ber of patients with a Fontan circulation and many, if not all,
will eventually experience failure of their Fontan circulation.
Results of VADs applied to failing univentricular circulation are
encouraging, and currently VAD support of the failing Fontan
as a bridge to transplantation is a viable strategy. However,
none of these devices have been designed to address the
majority of patients who have a Fontan circulation with pre-
dominantly right-sidedfailure. Novel devices designed to
support the low pressure, high flow cavopulmonary circulation
are under development, but have not yet been tested in vivo.
These devices offer hope of a fully implantable pump for
cavopulmonary support of the failing Fontan as destination
therapy.
5. Expert commentary
As the population of patients with a Fontan circulation surviv-
ing into adulthood continues to increase, clinicians will face
the clinical challenge represented by Fontan failure ever more
frequently. The current mainstay of therapy is heart transplan-
tation, but waiting times are long, donor supply is limited,
waiting list mortality is high, and we are often replacing the
heart in patients who have preserved systolic function.
Furthermore, heart transplantation substitutes one terminal
disease for another and does not offer these patients a chance
at normal life expectancy. The results of VAD use in patients
with a univentricular circulation are indeed encouraging, sug-
gesting that they are suitable for supporting patients with
Fontan failure who are deteriorating while awaiting transplan-
tation. However, the currently available technology is not well
suited to this application, being designed for support of the
systemic circulation, while more than half of all patients with
Fontan failure actually have preserved ventricular function.
Although the use of these devices to support the pulmonary
circulation has been reported, as they are designed for high
pressure environment, they are not optimized for this situa-
tion, unnecessarily increasing the risk of complications such as
pulmonary hypertension, venous collapse, obstruction of flows
and hemolysis.
We believe that a fully implantable low-pressure, high-
volume pump inserted into the Fontan pathway as cavopul-
monary support for use as destination therapy represents
thefutureofmanagingthefailingFontan.Thelowpower
requirements of this type of pump means that a long-life
battery, similar to that of a permanent pacemaker, should
be feasible, allowing intermittent replacement of a subcuta-
neous power supply. Alternatively, the advent of transcuta-
neous charging may eliminate the need for battery
replacement entirely. Allowing the device to become fully
implantable is a key to reducing the risk of infections. The
new pump designs are encouraging, especially that of
Rodefeldsgroup[63,64], which promotes a complex flow
pattern from the IVC and SVC into both pulmonary arteries.
Most impressively, the pump improves Fontan efficiency
even when stationary, minimizing the risk posed by device
failure, and providing a unique opportunity to more easily
conduct weaning trials. This is a key feature for the totally
implantable device to be successful as destination therapy
device failure must not result in circulatory obstruction. The
final major challenge which needs to be addressed is the
456 E. BURATTO ET AL.
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
risk of thrombogenicity, as the Fontan circulation is already
prone to thrombosis. However, devices such as Rodefelds
design, which improves blood streaming and reduces tur-
bulence at the TPCP, may go some way to mitigating this
risk.
As exciting as these technologies are, they remain a
long way from clinical practice, as none has yet been
tested even in animal models. Nevertheless, the growing
population of children and adults with a failing Fontan
circulation creates an urgent need for such devices,
which will no doubt expedite their development. A gen-
eration of children is relying on the community of cardiac
surgeons to provide them with a more certain future; a
fully implantable pump for cavopulmonary support
appears to be the way forward.
6. Five-year view
Given the promising results reported so far, in the next 5 years
we are likely to see increased use of current generation VADs
to support the failing Fontan circulation. It can be expected
that in the majority of cases these devices will be used as a
bridge to transplantation, as none of the devices are suitable
for destination therapy. Hence, heart transplantation will
remain the mainstay therapy for management of the failing
univentricular circulation, with VAD increasingly playing a
supporting role.
Testing of new-generation devices can be expected to
progress from the current in vitro experiments to animal test-
ing. It is possible, however, that toward the end of the next
5 years period we may see the first human trials of these novel
cardiopulmonary assist devices. While these developments are
exciting, the goal of a totally implantable cavopulmonary
support device as destination therapy appears to be more
than 5 years away from realization.
Key issues
As the population of Fontan patients increases, Fontan fail-
ure will be an increasingly common problem.
Current VADs are designed to support the systemic ventri-
cle. They are high-pressure pumps.
VADs have promising results for the support of the failing
Fontan circulation as a bridge to transplantation.
Most patients have right sided failurewith preserved sys-
tolic function. These patients would benefit from a low
powered pump for cavopulmonary support.
While novel low-pressure pumps for cavopulmonary sup-
port are under development, currently they remain at the
stage of in vitro testing.
The goal is a totally implantable pump for cavopulmonary
support with prolonged battery life which can be used as
destination therapy.
Funding
This paper was not funded.
Declaration of interest
E. Buratto is a recipient of a Reg Worcester Scholarship from the Royal
Australasian College of Surgeons and a Post Graduate scholarship from the
National Health and Medical Research Council (APP1134340). The authors
have no other relevant affiliations or financial involvement with any organi-
zation or entity witha financial interest in or financial conflict with the subject
matter or materials discussed in the manuscript apart from those disclosed.
References
Papers of special note have been highlighted as either of interest ()orof
considerable interest (••) to readers.
1. Rychik J. The relentless effects of the Fontan paradox. Semin
Thorac Cardiovasc Surg Pediatr Card Surg Annu. 2016;19:3743.
2. Jaquiss RDB, Aziz H. Is four stage management the future of uni-
ventricular hearts? Destination therapy in the young. Semin Thorac
Cardiovasc Surg Pediatr Card Surg Annu. 2016;19(1):5054.
•• Excellent review of Fontan physiology and potential role of
mechanical assist devices as destination therapy.
3. Gewillig M, Brown SC. The Fontan circulation after 45 years: update
in physiology. Heart. 2016;102:10811086.
4. dUdekem Y, Iyengar AJ, Galati JC, et al. Redefining expectations of
long-term survival after the Fontan procedure: twenty-five years of
follow-up from the entire population of Australia and New Zealand.
Circulation. 2014;130:S32S38.
5. dUdekem Y, Xu MY, Galati JC, et al. Predictors of survival after
single-ventricle palliation: the impact of right ventricular domi-
nance. J Am Coll Cardiol. 2012;59:11781185.
6. Hirsch JC, Goldberg C, Bove EL, et al. Fontan operation in the
current era: a 15-year single institution experience. Ann Surg.
2008;248:402410.
7. Giannico S, Hammad F, Amodeo A, et al. Clinical outcome of 193
extracardiac Fontan patients: the first 15 years. J Am Coll Cardiol.
2006;47:20652073.
8. Pundi KN, Johnson JN, Dearani JA, et al. 40-year follow-up after the
Fontan operation: long-term outcomes of 1,052 patients. J Am Coll
Cardiol. 2015;66:17001710.
9. Iyengar AJ, Sharma VJ, dUdekem Y, et al. Aortic arch and pulmon-
ary artery reconstruction during heart transplantation after failed
Fontan procedure. Interact Cardiovasc Thorac Surg. 2014;18:693
694.
10. Iyengar AJ, Sharma VJ, Weintraub RG, et al. Surgical strategies to
facilitate heart transplantation in children after failed univentricular
palliations: the role of advanced intraoperative surgical prepara-
tion. Eur J Cardiothorac Surg. 2014;46:480485.
11. Shi WY, Yong MS, McGiffin DC, et al. Heart transplantation in
Fontan patients across Australia and New Zealand. Heart.
2016;102:11201126.
•• Important paper demonstrating the outcomes of transplanta-
tion in the failing Fontan circulation.
12. Pundi KN, Pundi K, Driscoll DJ, et al. Heart transplantation after
Fontan: results from a surgical Fontan cohort. Pediatr Transplant.
2016;20:10871092.
13. Alsoufi B, Mahle WT, Manlhiot C, et al. Outcomes of heart trans-
plantation in children with hypoplastic left heart syndrome pre-
viously palliated with the Norwood procedure. J Thorac Cardiovasc
Surg. 2016;151:167174.
14. Shi WY, Rouse M, Weintraub RG, et al. Predictors of outcomes in
children awaiting heart transplantation: an experience from a
National Paediatric Heart Transplantation Programme. Eur J
Cardiothorac Surg. 2016;49:17111718.
15. Zafar F, Castleberry C, Khan MS, et al. Pediatric heart transplant
waiting list mortality in the era of ventricular assist devices. J Heart
Lung Transplant. 2015;34:8288.
16. Morales DLS, Almond CSD, Jaquiss RDB, et al. Bridging children of
all sizes to cardiac transplantation: the initial multicenter North
American experience with the Berlin Heart EXCOR ventricular assist
device. J Heart Lung Transplant. 2011;30:18.
EXPERT REVIEW OF MEDICAL DEVICES 457
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
17. Kantor PF, Redington AN. Pathophysiology and management of
heart failure in repaired congenital heart disease. Heart Fail Clin.
2010;6:497506.
18. Gewillig M, Brown SC, Eyskens B, et al. The Fontan circulation: who
controls cardiac output? Interact Cardiovasc Thorac Surg.
2010;10:428433.
19. Chowdhury UK, Airan B, Sharma R, et al. Univentricular repair in
children under 2 years of age: early and midterm results. Heart
Lung Circ. 2001;10:313.
20. Elder RW, Wu FM. Clinical approaches to the patient with a failing
Fontan procedure. Curr Cardiol Rep. 2016;18:44.
21. Kirklin JK, Naftel DC, Pagani FD, et al. Seventh INTERMACS annual
report: 15,000 patients and counting. J Heart Lung Transplant.
2015;34:14951504.
22. Tarzia V, Di Giammarco G, Di Mauro M, et al. From bench to bed-
side: can the improvements in left ventricular assist device design
mitigate adverse events and increase survival? J Thorac Cardiovasc
Surg. 2016 Jan;151:213217.
23. Tarzia V, Buratto E, Bortolussi G, et al. Hemorrhage and thrombosis
with different LVAD technologies: a matter of flow? Ann
Cardiothorac Surg. 2014;3:582584.
24. Tarzia V, Buratto E, Gallo M, et al. Implantation of the HeartWare
HVAD: from full sternotomy to less invasive techniques. Ann
Cardiothorac Surg. 2014;3:535537.
25. Slaughter MS, Rogers JG, Milano CA, et al. Advanced heart failure
treated with continuous-flow left ventricular assist device. N Engl J
Med. 2009;361:22412251.
26. Adachi I, Burki S, Zafar F, et al. Pediatric ventricular assist devices. J
Thorac Dis. 2015;7:21942202.
27. Fraser CD Jr, Jaquiss RD, Rosenthal DN, et al. Prospective trial of a
pediatric ventricular assist device. N Engl J Med. 2012;367:532541.
28. Rossano JW, Lorts A, VanderPluym CJ, et al. Outcomes of pediatric
patients supported with continuous-flow ventricular assist devices:
a report from the Pediatric Interagency Registry for Mechanical
Circulatory Support (PediMACS). J Heart Lung Transplant.
2016;35:585590.
29. Miera O, Kirk R, Buchholz H, et al. A multicenter study of the
HeartWare ventricular assist device in small children. J Heart Lung
Transplant. 2016;35:679681.
30. Rossano J, Villa C, Konstantinov I. Patient selection for ventricular
assist devices. In : Conway J, Lorts A, Schweiger M, Eds., ISHLT
Monograph Series: Pediatric Ventricular Assist Devices, Kirklin
Institute for Research in Surgical Outcomes, 2017, p 16.
31. Blume ED, Rosenthal DN, Rossano JW, et al. Outcomes of children
implanted with ventricular assist devices in the United States: first
analysis of the Pediatric Interagency Registry for Mechanical
Circulatory Support (PediMACS). J Heart Lung Transplant.
2016;35:578584.
•• Largest reported experience of VADs in children.
32. Rosenthal DN, Almond CS, Jaquiss RD, et al. Adverse events in children
implanted with ventricular assist devices in the United States: data from
the Pediatric Interagency Registry for Mechanical Circulatory Support
(PediMACS). J Heart Lung Transplant. 2016;35:569577.
33. Matsuda H, Taenaka Y, Ohkubo N, et al. Use of a paracorporeal
pneumatic ventricular assist device for postoperative cardiogenic
shock in two children with complex cardiac lesions. Artif Organs.
1988;12:423430.
34. Frazier OH, Gregoric ID, Messner GN. Total circulatory support with
an LVAD in an adolescent with a previous Fontan procedure. Tex
Heart Inst J. 2005;32:402404.
35. Nathan M, Baird C, Fynn-Thompson F, et al. Successful implantation
of a Berlin heart biventricular assist device in a failing single
ventricle. J Thorac Cardiovasc Surg. 2006;131:14071408.
36. Newcomb AE, Negri JC, Brizard CP, et al. Successful left ventricular
assist device bridge to transplantation after failure of a fontan
revision. J Heart Lung Transplant. 2006;25:365367.
37. Calvaruso DF, Ocello S, Salviato N, et al. Implantation of a Berlin
Heart as single ventricle by-pass on Fontan circulation in univen-
tricular heart failure. Asaio J. 2007;53:e1e2.
38. Chu MWA, Sharma K, Tchervenkov CI, et al. Berlin Heart ventricular
assist device in a child with hypoplastic left heart syndrome. Ann
Thorac Surg. 2007;83:11791181.
39. Prêtre R, Häussler A, Bettex D, et al. Right-sided univentricular
cardiac assistance in a failing Fontan circulation. Ann Thorac Surg.
2008;86:10181020.
40. Russo P, Wheeler A, Russo J, et al. Use of a ventricular assist device
as a bridge to transplantation in a patient with single ventricle
physiology and total cavopulmonary anastomosis. Paediatr
Anaesth. 2008;18:320324.
41. Cardarelli MG, Salim M, Love J, et al. Berlin heart as a bridge to
recovery for a failing Fontan. Ann Thorac Surg.
2009;87:943946.
42. Irving CA, Cassidy JV, Kirk RC, et al. Successful bridge to transplant
with the Berlin Heart after cavopulmonary shunt. J Heart Lung
Transplant. 2009;28:399401.
43. Pearce FB, Kirklin JK, Holman WL, et al. Successful cardiac trans-
plant after Berlin Heart bridge in a single ventricle heart: use of
aortopulmonary shunt as a supplementary source of pulmonary
blood flow. J Thorac Cardiovasc Surg. 2009;137:e40e42.
44. Miera O, Potapov EV, Redlin M, et al. First experiences with the
HeartWare ventricular assist system in children. Ann Thorac Surg.
2011;91:12561260.
45. VanderPluym CJ, Rebeyka IM, Ross DB, et al. The use of ventricular
assist devices in pediatric patients with univentricular hearts. J
Thorac Cardiovasc Surg. 2011;141:588590.
46. Mackling T, Shah T, Dimas V, et al. Management of single-ventricle
patients with Berlin Heart EXCOR ventricular assist device: single-
center experience. Artif Organs. 2012;36:555559.
47. Brancaccio G, Gandolfo F, Carotti A, et al. Ventricular assist device
in univentricular heart physiology. Interact Cardiovasc Thorac Surg.
2013;16:568569.
48. Sanders DB, Sowell SR, Park SS, et al. The failing Fontan: whats
NEXT. . .? Perfusion. 2014;29:8993.
49. Valeske K, Yerebakan C, Mueller M, et al. Urgent implantation of the
Berlin Heart EXCOR biventricular assist device as a total artificial
heart in a patient with single ventricle circulation. J Thorac
Cardiovasc Surg. 2014;147:17121714.
50. Weinstein S, Bello R, Pizarro C, et al. The use of the Berlin Heart
EXCOR in patients with functional single ventricle. J Thorac
Cardiovasc Surg. 2014;147:6975.
•• Largest experience of VAD support of patients with single
ventricle physiology.
51. Arnaoutakis GJ, Blitzer D, Fuller S, et al. Mechanical circulatory
support as bridge to transplantation for the failing single ventricle.
Ann Thorac Surg. 2017;103(1):193197.
52. Niebler RA, Shah TK, Mitchell ME, et al. Ventricular assist device in
single-ventricle heart disease and a superior cavopulmonary ana-
stomosis. Artif Organs. 2016;40:180184.
53. Elder RW, McCabe NM, Hebson C, et al. Features of portal hyper-
tension are associated with major adverse events in Fontan
patients: the VAST study. Int J Cardiol. 2013;168:37643769.
54. Griffiths ER, Kaza AK, Wyler von Ballmoos MC, et al. Evaluating
failing Fontans for heart transplantation: predictors of death. Ann
Thorac Surg. 2009 Aug;88:5584.
55. Rodefeld MD, Frankel SH, Giridharan GA. Cavopulmonary assist:
(em)powering the univentricular fontan circulation. Semin Thorac
Cardiovasc Surg Pediatr Card Surg Annu. 2011;14:4554.
•• Excellent review of the role for cavopulmonary support in the
Fontan circulation.
56. De Leval MR. The Fontan circulation: a challenge to William
Harvey? Nat Clin Pract Cardiovasc Med. 2005;2:202208.
Excellent review of Fontan physiology.
57. Wei X, Sanchez PG, Liu Y, et al. Mechanical circulatory support of a
univentricular Fontan circulation with a continuous axial-flow
pump in a piglet model. Asaio J. 2015;61:196201.
58. Derk G, Laks H, Biniwale R, et al. Novel techniques of mechanical
circulatory support for the right heart and Fontan circulation. Int J
Cardiol. 2014;176:828832.
458 E. BURATTO ET AL.
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
59. Riemer RK, Amir G, Reichenbach SH, et al. Mechanical support of
total cavopulmonary connection with an axial flow pump. J Thorac
Cardiovasc Surg. 2005;130:351354.
60. Wang R, Lacour-Gayet FG, Lanning CJ, et al. Initial experience with
the development and numerical and in vitro studies of a novel low-
pressure artificial right ventricle for pediatric Fontan patients. Asaio
J. 2006;52:682692.
61. Zhu J, Kato H, Fu YY, et al. Cavopulmonary support with a
microaxial pump for the failing Fontan physiology. Asaio J.
2015;61:4954.
62. Gandolfo F, Brancaccio G, Donatiello S, et al. Mechanically
assisted total cavopulmonary connection with an axial flow
pump: computational and in vivo study. Artif Organs.
2016;40:4349.
63. Rodefeld MD, Coats B, Fisher T, et al. Cavopulmonary assist for the
univentricular Fontan circulation: von Kármán viscous impeller
pump. J Thorac Cardiovasc Surg. 2010;140:529536.
Promising example of a novel pump for cavopulmonary
support.
64. Giridharan GA, Koenig SC, Kennington J, et al. Performance evalua-
tion of a pediatric viscous impeller pump for Fontan cavopulmon-
ary assist. J Thorac Cardiovasc Surg. 2013;145:249257.
Promising example of a novel pump for cavopulmonary support.
65. Lacour-Gayet FG, Lanning CJ, Stoica S, et al. An artificial right
ventricle for failing fontan: in vitro and computational study. Ann
Thorac Surg. 2009;88:170176.
66. Throckmorton AL, Kapadia J, Madduri D. Mechanical axial flow
blood pump to support cavopulmonary circulation. Int J Artif
Organs. 2008;31:970982.
67. Bhavsar SS, Kapadia JY, Chopski SG, et al. Intravascular mechanical
cavopulmonary assistance for patients with failing Fontan physiol-
ogy. Artif Organs. 2009;33:977987.
68. Chopski SG, Fox CS, Riddle ML, et al. Pressure-flow experimental
performance of new intravascular blood pump designs for Fontan
patients. Artif Organs. 2016;40:233242.
69. Kafagy DH, Dwyer TW, McKenna KL, et al. Design of axial blood
pumps for patients with dysfunctional fontan physiology: computa-
tional studies and performance testing. Artif Organs. 2015;39:3442.
EXPERT REVIEW OF MEDICAL DEVICES 459
Downloaded by [The University Of Melbourne Libraries] at 17:37 06 November 2017
... 15 Needless to say, the configuration of VAD support must be tailored to the mode of Fontan failure and the procedure must be performed by a surgeon with expertise in managing patients with univentricular circulation. [16][17][18] In particular, patients with preserved ventricular function often have a substantial burden of aortopulmonary collaterals and higher pulmonary vascular resistance, and appear to have increased mortality compared with patients with impaired function. 4 Multiorgan dysfunction, especially renal and liver failure and plastic bronchitis, commonly occurs in the failing Fontan circulation. ...
... We and others have previously emphasized this approach as a bridge to candidacy for heart transplantation in patients with failing Fontan circulation and preserved single ventricle function. 16,21,22 Yet, the current world experience is still limited to a handful of patients. Although a combined heart-lung-liver transplantation is technically feasible, 23 the need for such procedure ...
... 42,180,184,[193][194][195][196][197][198][199][200][201][202][203] There are also case reports of DT SVAD support for Fontan patients, 204 as well as VAD support of the pulmonary circulation. 205 Additionally, SVAD support in Fontan patients as bridge to combined multi-organ (heart-liver, heart-kidney) transplant have not yet been reported. ...
... With reference to the classification of VAD support in Fontan patients by Buratto et al. the application of a singlechambered TAH can be discussed for both scenarios [28]. Patients with a systolic ventricular dysfunction develop a raised venous pressure. ...
Article
Full-text available
An in-vitro study was conducted to investigate the general feasibility of using only one pumping chamber of the SynCardia total artificial heart (TAH) as a replacement of the single ventricle palliated by Fontan circulation. A mock circulation loop was used to mimic a Fontan circulation. The combination of both ventricle sizes (50 and 70 cc) and driver (Freedom Driver and Companion C2 Driver) was investigated. Two clinical relevant scenarios (early Fontan; late Fontan) as derived from literature data were set up in the mock loop. The impact of increased transpulmonary pressure gradient, low atrial pressure, and raised central venous pressure on cardiac output was studied. From a hemodynamic point, the single-chambered TAH performed sufficiently in the setting of the Fontan circulation. Increased transpulmonary pressure gradient, from ideal to pulmonary hypertension, decreased the blood flow in combinations by almost 2 L/min. In the early Fontan scenario, a cardiac output of 3–3.5 L/min was achieved using the 50 cc ventricle, driven by the Companion C2 Driver. Even under pulmonary hypertension, cardiac outputs greater than 4 L/min could be obtained with the 70 cc pump chamber in the late Fontan scenario. In the clinically relevant Fontan scenarios, implementation of the single chambered TAH performed successfully from a hemodynamic point of view. The replacement of the failing univentricular heart by a single chamber of the SynCardia TAH may provide an alternative to a complex biventricular repair procedure or ventricular support in Fontan patients.
... Most reports rely on assist support for the systemic ventricle or total artificial heart in Fontan circulation. 3,4 In contrary to patients with chronic heart failure, Fontan patients with PLE present with a discrepancy between poor clinical condition and preserved ejection fraction of the single ventricle. Prêtre ...
Article
Full-text available
Fontan patients with protein‐losing enteropathy (PLE) represent poor candidates for cardiac transplantation due to end‐organ injury and severely impaired clinical condition. Ventricular assist device (VAD) therapy has evolved as a promising bridge to transplant strategy improving quality of life and survival on the waiting list. However, VAD therapy for the Fontan circulation remains challenging. For Fontan patients with preserved ventricular function implantation of a right ventricular assist device (RVAD) has been described by Prêtre et al as bridge to transplant. We present the second case of RVAD support in a Fontan patient with PLE.
Article
Full-text available
Background The number of single ventricle patients undergoing Fontan palliation and surviving to adulthood worldwide has steadily increased in recent years. Nevertheless, the Fontan circulation is destined to fail. Ultimately, heart transplantation (HTx) remains the definitive treatment option. Due a shortage of organs, mechanical circulatory support in the form of ventricular assist devices (VADs) is widely used to bridge heart failure patients to HTx, but these devices have been mainly developed to address the needs of normal anatomies. A novel venous cannula has been developed as part of the EXCOR® VAD to provide subpulmonary support in these patients. Its clinical application is investigated in the “Registry to Assess the Safety and Feasibility of the Subpulmonary Support with the Novel Venous Cannula in Patients with Failing/Absence of the Right Heart” (RegiVe study, NCT04782232). Methods RegiVe is a multicenter, international, observational, prospective, non-randomized registry aiming to collect the routine clinical data of up to 20 patients. The primary endpoints address device performance and safety, while the secondary endpoints target organ status and overall safety (according to the Interagency Registry for Mechanically Assisted Circulatory Support – INTERMACS – definitions). Data analysis will be performed by means of descriptive statistics. Results RegiVe has received the favorable opinion of an independent ethics committee and enrollment has recently started. Conclusion RegiVe is the first study evaluating the use of a medical device specifically developed for subpulmonary support of failing Fontan patients. The study will provide important insight and further information on this cohort and help to improve a dedicated VAD strategy.
Article
Full-text available
The Fontan operation was introduced in 1968. For congenital malformations where biventricular repair is not suitable, the Fontan procedure has provided a long-term palliation strategy with improved outcome compared to the initially developed procedures. Despite these improvements, several complications merely as a result of a failing Fontan circulation (including myocardial dysfunction, arrhythmias, increased pulmonary vascular resistance, protein losing enteropathy, hepatic dysfunction, plastic bronchitis and thrombo-embolism) will limit life-expectancy in this patient cohort. This review provides an overview of the most common complications of the Fontan circulation and the currently available treatment options.
Article
Full-text available
Advances in surgical techniques and outpatient cardiological care have led to a growing population of pediatric patients surviving well into adulthood with previous single ventricle palliation. As survival continues to improve, subsequent increases in the number of patients with single ventricle physiology listed for heart transplants have resulted. Some of these patients require mechanical circulatory support (MCS) as a bridge to transplant, although establishing successful MCS in these complex patients remains challenging. Only limited published data exist describing the perioperative anesthetic management and key considerations dedicated to patients with failing single ventricle physiology presenting for ventricular assist devices (SVAD). This clinical review aims to provide a focused evaluation of the vital perioperative considerations encountered in this novel population.
Article
Progressive aortic regurgitation (AR) is a common complication in patients supported with continuous flow ventricular assist devices (VADs). The risk of neo-AR is likely to be particularly high in patients with univentricular hearts, due to the lack of fibrous support for the neo-aortic valve. Previously described techniques for addressing neo-AR in this setting have required cardiopulmonary bypass and cross clamping, or ligation of the neo aortic root. We present a simple technique of external partial annuloplasty of the neo-aortic valve, which can be performed without the need for cardiopulmonary bypass or cross clamping.
Article
There is mounting recognition that some of the most urgent problems of adult congenital heart disease (ACHD) are the prevention, diagnosis, and management of heart failure (HF). Recent expert consensus and position statements not only emphasize a specific and pressing need to tackle HF in ACHD (ACHD-HF) but also highlight the difficulty of doing so given a current sparsity of data. Some of the challenges will be addressed by this review. The authors are from 3 different centres; each centre has an established subspeciality ACHD-HF clinic and is able to provide heart transplant, multiorgan transplant, and mechanical support for patients with ACHD. Appropriate care of this complex population requires multidisciplinary ACHD-HF teams evaluate all possible treatment options. The risks and benefits of nontransplant ACHD surgery, percutaneous structural and electrophysiological intervention, and ongoing conservative management must be considered alongside those of transplant strategies. In our approach, advanced care planning and palliative care coexist with the consideration of advanced therapies. An ethos of shared decision making, guided by the patient's values and preferences, strengthens clinical care, but requires investment of time as well as skilled communication. In this review, we aim to offer practical real-world advice for managing these patients, supported by scientific data where it exists.
Article
Full-text available
The Fontan operation was first performed in 1968. Since then, this operation has been performed on thousands of patients worldwide. Results vary from very good for many decades to very bad with a pleiad of complications and early death. A good understanding of the physiology is necessary to further improve results. The Fontan connection creates a critical bottleneck with obligatory upstream congestion and downstream decreased flow; these two features are the basic cause of the majority of the physiologic impairments of this circulation. The ventricle, while still the engine of the circuit, cannot compensate for the major flow restriction of the Fontan bottleneck: the suction required to compensate for the barrier effect cannot be generated, specifically not in a deprived heart. Except for some extreme situations, the heart therefore no longer controls cardiac output nor can it significantly alter the degree of systemic venous congestion. Adequate growth and development of the pulmonary arteries is extremely important as pulmonary vascular impedance will become the major determinant of Fontan outcome. Key features of the Fontan ventricle are early volume overload and overgrowth, but currently chronic preload deprivation with increasing filling pressures. A functional decline of the Fontan circuit is expected and observed as pulmonary vascular resistance and ventricular filling pressure increase with time. Treatment strategies will only be successful if they open up or bypass the critical bottleneck or act on immediate surroundings (impedance of the Fontan neoportal system, fenestration, enhanced ventricular suction).
Article
Full-text available
For the child born with a functionally univentricular heart, multi-stage surgical palliation culminating in the Fontan operation is now well established as the standard of care. Though this is an effective approach, there are several inherent disadvantages to the Fontan circulation: subnormal resting cardiac output, impaired exercise capacity, increased risk of thromboembolism, and increased risk of arrhythmia. Perhaps most importantly, the long-term deleterious effects of substantial systemic venous hypertension, which is necessary to "drive" blood through the pulmonary vascular bed, include progressive dysfunction of other organ systems, particularly renal, lymphatic, gastrointestinal, and hepatic systems. When such dysfunction is advanced, heart transplantation or even heart-liver transplantation may be a reasonable option. However, because the syndrome seems to be increasingly widespread and there is already a significant donor shortage, alternative solutions are required. Because many patients with "failing Fontan" physiology have preserved systemic ventricular function, application of a systemic ventricular assist device is unlikely to be effective. However, for such patients, a right-sided sub-pulmonary ventricular assist device is an intellectually appealing solution. Several such devices have been proposed or are in varying stages of evaluation. The lack of economic incentive for development of right-sided pumps may be partially ameliorated both by recognition of the size of the cohort of Fontan patients now surviving into adulthood as well as by the increasing recognition of important right-sided heart failure in adults with biventricular hearts supported only with durable left-sided ventricular assist devices.
Article
Full-text available
The Fontan operation is the anticipated palliative strategy for children born with single-ventricle type of congenital heart disease. As a result of important circulatory limitations, a series of end-organ complications are now increasingly recognized. Elevated central venous pressure and impaired cardiac output are the hallmarks of cavo-pulmonary flow, which result in a cascade of pathophysiological consequences. The Fontan circulation likely impacts all organ systems in an indolent and relentless manner, with progressive decline in functionality likely to occur in many. Liver fibrosis, altered bone density, decreased muscle mass, renal dysfunction, lymphatic insufficiency, and a host of other conditions are present. Standardized screening and evaluation of survivors as they grow through childhood and beyond is indicated and can be facilitated through dedicated multidisciplinary clinical programs. Invasive assessment at specific milestones can provide important actionable information to optimize individual status. More detailed characterization and understanding of these end-organ complications is necessary to contribute to the goal of achieving a normal duration and quality of life for these unique individuals.
Article
Background: Circulatory failure necessitating cardiac transplantation will ultimately develop in many patients with functional single-ventricle physiology. Interest in the use of mechanical circulatory support (MCS) in this population is growing. Methods: This was a retrospective case series of patients with functional single-ventricle physiology who underwent MCS with a ventricular assist device or a total artificial heart as a bridge to cardiac transplantation between January 2006 and December 2014. Baseline demographics, intraoperative data, postoperative complications, and outcome data were collected from the medical record. Results: MCS was used in 5 patients: HeartWare ventricular assist device (HeartWare International, Framingham, MA) in 1 patient, SynCardia total artificial heart (SynCardia Systems, Tucson, AZ) in 1, Thoratec Paracorporeal ventricular assist device (Thoratec Corp, Pleasanton, CA) in 1, and the Berlin Heart EXCOR (Berlin Heart Inc, The Woodlands, TX) in 2. The mean age at MCS was 12 ± 8 years. There were 2 early deaths at 12 and 28 days after MCS: 1 patient died of multiorgan system failure and 1 of neurologic injury. Overall, neurologic complications occurred in 3 patients (60%), and 1 patient (20%) required renal replacement therapy. Three patients (60%) underwent successful cardiac transplantation. The median time on the waiting list was 59 days (interquartile range, 18 to 126 days), and the median duration of MCS was 60 days (interquartile range, 28 to 93 days). At the time of transplant, all 3 patients were ambulatory, without the need for mechanical ventilation, and end-organ dysfunction had resolved. The 3 patients who received transplants were discharged from the hospital and were alive at an average follow-up of 9 ± 14 months. Conclusions: MCS can be successfully used as a bridge to transplantation in patients with a failing single-ventricle circulation. Use of MCS can allow for resolution of end-organ dysfunction and rehabilitation, leading to improved outcomes in this difficult population.
Article
We performed a retrospective review of outcomes after heart transplantation during long-term follow-up of a surgical cohort of 1138 Fontan patients who were followed at the Mayo Clinic. Follow-up information was obtained from medical records and a clinical questionnaire that was mailed to patients not known to be deceased at the initiation of the study. Forty-four of 1138 Fontan patients with initial or subsequent evaluation at Mayo had cardiac transplantation between 1988 and 2014 (mean age at transplantation was 23.2 ± 12 yr, median was 19.8 yr; mean interval between Fontan and transplantation was 13.0 ± 7.7 yr, median was 13.1 yr). Two patients had combined organ transplantation (one heart-lung, one heart-liver). Twelve of the 44 (27%) patients had PLE prior to transplantation. There was no difference in post-bypass Fontan pressures or incidence of late reoperations for AVV repair/replacement between transplanted and non-transplanted patients. There were 16 (36%) deaths in the transplantation cohort; seven occurred within 30 days of transplantation. Overall one, five, 10, and 15 yr post-transplantation survival was 80%, 72%, 69%, and 55%, respectively. Although this is a challenging group of patients, intermediate-term results suggest that cardiac transplantation remains a reasonable option for patients with a failed Fontan circulation.
Article
Background: Ventricular assist devices (VADs) have been used in children on an increasing basis in recent years. One-year survival rates are now >80% in multiple reports. In this report we describe adverse events experienced by children with durable ventricular assist devices, using a national-level registry (PediMACS, a component of INTERMACS) METHODS: PediMACS is a national registry that contains clinical data on patients who are <19 years of age at the time of VAD implantation. Data collection concludes at the time of VAD explantation. All FDA-approved devices are included. PediMACS was launched on September 1, 2012, and this report includes all data from launch until August 2014. Adverse events were coded with a uniform, pre-specified set of definitions. Results: This report comprises data from 200 patients with a median age of 11 years (range 11 days to 18 years), and total follow-up of 783 patient-months. The diagnoses were cardiomyopathy (n = 146, 73%), myocarditis (n = 17, 9%), congenital heart disease (n = 35, 18%) and other (n = 2, 1%). Pulsatile-flow devices were used in 91 patients (45%) and continuous-flow devices in 109 patients (55%). Actuarial survival was 86% at 6 months. There were 418 adverse events reported. The most frequent events were device malfunction (n = 79), infection (n = 78), neurologic dysfunction (n = 52) and bleeding (n = 68). Together, these accounted for 277 events, 66% of the total. Although 38% of patients had no reported adverse event and 16% of patients had ≥5 adverse events. Adverse events occurred at all time-points after implantation, but were most likely to occur in the first 30 days. For continuous-flow devices, there were broad similarities in adverse event rates between this cohort and historic rates from the INTERMACS population. Conclusions: In this study cohort, the overall rate of early adverse events (within 90 days of implantation) was 86.3 events per 100 patient-months, and of late adverse events it was 20.4 events per 100 patient-months. The most common adverse events in recipients of pulsatile VADs were device malfunction, neurologic dysfunction, bleeding and infection. For continuous-flow VADs, the most common adverse events were infection, bleeding, cardiac arrhythmia, neurologic dysfunction and respiratory failure. Compared with an adult INTERMACS cohort, the overall rate and distribution of adverse events appears similar.
Article
Background: Continuous-flow (CF) ventricular assist devices (VADs) have largely replaced pulsatile-flow VADs in adult patients. However, there are few data on CF VADs among pediatric patients. In this study we aimed to describe the overall use, patients' characteristics and outcomes of CF VADs in this population. Methods: The Pediatric Interagency Registry for Mechanical Circulatory Support (PediMACS) is a national registry for U.S. Food and Drug Adminstration (FDA)-approved VADs in patients <19 years of age. Patients undergoing placement of durable CF VADs between September 2012 and June 2015 were included and outcomes were compared with those of adults from the Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS). Results: CF VADs were implanted in 109 patients at 35 hospitals. The median age at implantation was 15 years (2.8 to 18.9 years) and median weight was 62 kg (range 16 to 141 kg). The underlying disease was cardiomyopathy in 89 (82%) patients. The INTERMACS level at time of implant was Level 1 in 20 (19%), Level 2 in 64 (61%) and Levels 3 to 7 in 21 (20%) patients. Most were implanted as LVADs (n = 102, 94%). Median duration of support was 2.3 months (range <1 day to 28 months). Serious adverse event rates were low, including neurologic dysfunction (early event rate 4.1 per 100 patient-months with 2 late events). Competing outcomes analysis at 6 months post-implant indicated 61% transplanted, 31% alive with device in place and 8% death before transplant. These outcomes compared favorably with the 3,894 adults supported with CF VADs as a bridge to transplant. Conclusions: CF VADs are commonly utilized in older children and adolescents, with excellent survival rates. Further study is needed to understand impact of patient and device characteristics on outcomes in pediatric patients.